1
|
Ge P, Wang J, Xie P, Hu Z. Exosomes in treating Eye Diseases: Targeting strategies. Life Sci 2025; 371:123598. [PMID: 40174670 DOI: 10.1016/j.lfs.2025.123598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 03/25/2025] [Accepted: 03/29/2025] [Indexed: 04/04/2025]
Abstract
Exosomes are lipid-based vesicles carrying bioactive molecules (nucleic acids, proteins, lipids) that mediate intercellular communication. Emerging research explores their potential as therapeutic delivery systems, with bioengineering approaches enhancing stability and efficacy for clinical translation. This review focuses on exosome applications in ocular diseases, particularly engineered targeting strategies, while addressing current limitations and future clinical prospects.
Collapse
Affiliation(s)
- Pengfei Ge
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jingfan Wang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Ping Xie
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Zizhong Hu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
2
|
XU K, HUANG Y, ZHAO R. [Research progress of peptide recognition-guided strategies for exosome isolation and enrichment]. Se Pu 2025; 43:446-454. [PMID: 40331609 PMCID: PMC12059989 DOI: 10.3724/sp.j.1123.2024.10015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Indexed: 05/08/2025] Open
Abstract
Exosomes are bilayered vesicles derived from living cells and bacteria that are loaded with abundant biomolecules, such as proteins and nucleic acids. As an important medium of remote cell communication, exosomes are closely related to the occurrence and development of a number of diseases, including those involving tumors and inflammation. The isolation and enrichment of exosomes in complex biosystems is greatly significant for the diagnosis, prognosis, and detection of diseases, as well as in molecular-mechanism research. However, exosomes are usually nanoscale size distribution and widely existed in complex biological samples, including blood, tissue fluids, and urine, which bring difficulties and challenges to the isolation and enrichment of exosomes. To address this issue, several methods based on the physical properties of exosomes have been developed. For example, exosomes can be obtained by ultracentrifugation at high centrifugal force based on density differences; they can also be isolated and enriched by size-exclusion chromatography and ultrafiltration based on size heterogeneity. Exosomes can also be separated in high yields but with low purities using commercial polymer-coprecipitation-based isolation kits. While the abovementioned methods can be used to isolate and enrich exosomes in a highly efficient manner, accurately distinguishing interfering particles, including protein aggregates and microvesicles, in biosystems is still difficult, resulting in the poor purity of exosome isolation and enrichment. Affinity ligands are widely used during the affinity isolation and enrichment of exosomes. Antibodies exhibit high selectivity and affinity; consequently exosomes can be captured highly selectively by exploiting specific antigen/antibody interactions. However, antibodies also have some limitations, including complex preparation procedures, high costs, and poor stability. Chemical affinity ligands, such as aptamers, peptides, and small molecules, are also widely used to isolate and enrich exosomes. As a kind of molecular recognition tool, peptides contain a variety of amino acids and exhibit many advantages, including good biocompatibility, low immunogenicity, and design flexibility. Solid-phase synthesis strategies have rapidly developed, thereby providing a basis for automated and large-scale peptide synthesis. Affinity peptides have been widely used to recognize and analyze target biomolecules in complex physiological environments in a highly selective manner. A series of protein-targeting peptides has been reported based on the biomolecular characteristics of exosomes. These affinity peptides can be specifically anchored onto highly enriched transmembrane proteins on exosome surfaces, thereby enabling the efficient and highly selective isolation and enrichment of exosomes in complex systems. Additionally, exosomes contain stable bilayer membranes consisting of abundant and diverse phospholipid molecules. The development of phospholipid-molecule-targeting peptides is expected to effectively eliminate interference from protein aggregates and other particles. In addition to differences in the compositions of phospholipids in biofilms, exosomes are smaller and more curved than apoptotic bodies and microvesicles. A series of affinity peptides capable of inducing and sensing high membrane curvatures are widely used to isolate and enrich exosomes. The tumor microenvironment can produce and release tumor-derived exosomes that are buried in a large number of normal cell-derived exosomes. Accordingly, pH-responsive peptides have been designed and modified based on the acidic environments of tumor-derived exosomes, which were accurately and tightly anchored via peptide insertion and folding. Focusing on the current status of exosome research, this paper introduces and summarizes current and widely used methods for isolating and enriching exosomes. Various exosome-targeting peptide-design and screening principles are introduced based on the characteristics and advantages of peptides. The applications of these peptides to the isolation and enrichment of exosomes are also summarized, thereby providing strong guidance for the efficient and highly selective isolation and enrichment of exosomes in complex life-related systems.
Collapse
|
3
|
Tang X, Ma C, Ren Y, Lv Y, He Y, Han L, Wu J. Revealing the Potential of Solamargine for Anti Metastasis and Angiogenesis Inhibition in Nasopharyngeal Carcinoma. J Inflamm Res 2025; 18:4879-4898. [PMID: 40224395 PMCID: PMC11992990 DOI: 10.2147/jir.s485244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 02/06/2025] [Indexed: 04/15/2025] Open
Abstract
Background Nasopharyngeal carcinoma (NPC) is a major global health issue, especially in Southeast Asia. Solamargine (SM), an alkaloid from natural plants, inhibits various cancer cells. This study evaluates SM's effects on invasion, migration, EMT markers, angiogenesis, and related pathways in the NPC-specific C666-1 cell line. Methods In vitro assays, including wound healing, Transwell invasion, Western blot, and tube formation, were used to assess SM's impact on C666-1 NPC and HUVEC cells. SM concentrations were 2 µM and 5 µM, with axitinib (4 µM) as the control. Network pharmacology and GO-KEGG enrichment analyses were conducted to explore SM's targets and mechanisms in NPC. Results SM significantly inhibited C666-1 NPC cell invasion and migration by reducing EMT markers Vimentin and Snail. In HUVEC cells, SM decreased viability, invasion, migration, and tube formation, likely through VEGF signaling inactivation, EZH2 inhibition, and miR-203a-3p upregulation. Network pharmacology and GO-KEGG analyses identified key targets and pathways, suggesting SM's anti-NPC effects through multiple mechanisms. Discussion SM inhibits NPC cell invasion and migration by regulating EMT, suppressing angiogenesis, and modulating key pathways. These findings highlight SM's potential as an anti-cancer agent for NPC and provide new insights into its mechanisms. Network pharmacology and GO-KEGG analysis further identify its therapeutic targets, offering valuable directions for future drug development.
Collapse
Affiliation(s)
- Xiaojuan Tang
- Central Laboratory, Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, 410006, People’s Republic of China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, People’s Republic of China
| | - Changju Ma
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, People’s Republic of China
| | - Yuan Ren
- Central Laboratory, Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, 410006, People’s Republic of China
| | - Yuan Lv
- Central Laboratory, Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, 410006, People’s Republic of China
| | - Yongheng He
- Department of Anorectal Surgery, Affiliated Hospital of Hunan Academy of Traditional Chinese Medicine, Changsha, 410006, People’s Republic of China
| | - Ling Han
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, People’s Republic of China
| | - Jingjing Wu
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, People’s Republic of China
| |
Collapse
|
4
|
Zhuo H, Zhang S, Wang H, Deng J, Zhang X. Gelatin methacryloyl @MP196/exos hydrogel induced neutrophil apoptosis and macrophage M2 polarization to inhibit periodontal bone loss. Colloids Surf B Biointerfaces 2025; 248:114466. [PMID: 39729702 DOI: 10.1016/j.colsurfb.2024.114466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 12/13/2024] [Accepted: 12/17/2024] [Indexed: 12/29/2024]
Abstract
OBJECTIVES Periodontitis is an inflammatory and destructive disease caused by dental plaque, which can result in the immune microenvironment disorders and loss of periodontal support tissue. In order to promote the restoration of local microenvironment stability, a functional biomaterial Gelatin methacryloyl @MP196/exos based on characteristics of disease occurrence is designed. METHODS Transmission electron microscopy, nanosight particle tracking analysis and western blot analysis were applied to prove the presence of exos in GelMA@MP196/exos. The swelling and degradation rates of GelMA@MP196/exos were evaluated. Cell proliferation, antibacterial ability and cellular uptake and intracellular internalization of exos were assessed in the study. Efferocytosis and M2 polarization of macrophages was estimated and the effects of GelMA@MP196/exos were proved in vivo. RESULTS GelMA@MP196/exos upregulated the expression of genes and proteins related to neutrophil apoptosis and promoted neutrophil apoptosis, macrophage M2 polarization, and efferocytosis. Furthermore, GelMA@MP196/exos exhibited significant antibacterial activity against Streptococcus gordonii, Fusobacterium nucleatum, and Porphyromonas gingivalis. GelMA@MP196/exos alleviated periodontitis and reduced alveolar bone loss in vivo in rat models. CONCLUSIONS GelMA@MP196/exos can serve as a potential strategy for the treatment of periodontitis. CLINICAL SIGNIFICANCE The main aim of periodontal therapy is to remove dental plaque and eliminate inflammation. However, some patients with low plaque scores and insufficient neutrophil clearance, resulting in poor responsiveness to periodontal therapy. Under the circumstances, local Application of drug that regulate the immune microenvironment had significance in controlling the progression of inflammation.
Collapse
Affiliation(s)
- Haiwei Zhuo
- Department of Periodontology, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China; Tianjin Medical University Institute of Stomatology, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China
| | - Shuting Zhang
- Department of Periodontology, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China; Tianjin Medical University Institute of Stomatology, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China
| | - Hongbo Wang
- Department of Periodontology, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China; Tianjin Medical University Institute of Stomatology, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China
| | - Jiayin Deng
- Department of Periodontology, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China; Tianjin Medical University Institute of Stomatology, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China.
| | - Xi Zhang
- Department of Periodontology, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China; Tianjin Medical University Institute of Stomatology, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China.
| |
Collapse
|
5
|
Liu Y, Zhou H, Pang P, Liu Y, Situ J, Wang J, He Y, Ye Q. Tremella polysaccharide microneedles loaded with magnetic dental pulp stem cell intracellular vesicles used for androgenic alopecia. Stem Cell Res Ther 2025; 16:161. [PMID: 40165226 PMCID: PMC11959820 DOI: 10.1186/s13287-025-04219-3] [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: 11/12/2024] [Accepted: 02/11/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Mesenchymal stem cell exosomes are the most extensively researched type of Extracellular vesicles (EVs) that offer novel avenues for hair regeneration. However, their use in the field of hair regeneration was limited by their poor production of exosomes. It has been discovered that intracellular vesicles (IVs), which are produced at a higher rate than exosomes, play a comparable biological purpose. As a result, we developed HTMI-MN, a microneedle that uses tremella, a type of Tremella polysaccharide (TPS), and hyaluronic acid (HA) as matrix materials. It has magnetic intracellular vesicles (Mag-IVs), which work in concert to treat androgenic alopecia (AGA) and encourage hair growth. METHODS After characterization of the Mag-IVs, we evaluated the effect on angiogenesis by scratch assay, angiogenesis assay, Western Blot and ELISA assay. In addition, we tested the protective effect of Human hair papillary cells (HHDPCs) by CCK-8 method, Western Blot and flow cytometry. Finally, the effects of tremella polysaccharide on M1/M2 polarization of macrophages were detected by fluorescence staining, Western Blot and flow cytometry. AGA model was established in vivo by DHT, and treatment was given by microneedle injection. RESULTS Our study found that Mag-IVs have greater power to promote angiogenesis and protect HHDPCs from apoptosis compared to other vesicles. Besides, tremella polysaccharide can make the transformation of macrophages to anti-inflammatory phenotype. Taken together, in vivo experiments showed that hair regeneration was faster in HTMI-MN-treated mice. CONCLUSION These results indicate that Mag-IVs and tremella polysaccharide can synergistically improve the hair microenvironment, which has a promising future for AGA treatment.
Collapse
Affiliation(s)
- Yonghao Liu
- Center of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Heng Zhou
- Center of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Pengcheng Pang
- Center of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ye Liu
- Center of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jieying Situ
- Center of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Junnan Wang
- Center of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yan He
- Institute of Regenerative and Translational Medicine, Tianyou Hospital of Wuhan University of Science and Technology, Wuhan, Hubei, 430064, China.
| | - Qingsong Ye
- Center of Regenerative Medicine, Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- School of Dentistry, The University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
6
|
Abbasi M, Aghamollaei H, Vaez A, Amani AM, Kamyab H, Chelliapan S, Jamalpour S, Zambrano-Dávila R. Bringing ophthalmology into the scientific world: Novel nanoparticle-based strategies for ocular drug delivery. Ocul Surf 2025; 37:140-172. [PMID: 40147816 DOI: 10.1016/j.jtos.2025.03.004] [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: 01/28/2025] [Revised: 03/03/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
The distinctive benefits and drawbacks of various drug delivery strategies to supply corneal tissue improvement for sense organs have been the attention of studies worldwide in recent decades. Static and dynamic barriers of ocular tissue prevent foreign chemicals from entering and inhibit the active absorption of therapeutic medicines. The distribution of different medications to ocular tissue is one of the most appealing and demanding tasks for investigators in pharmacology, biomaterials, and ophthalmology, and it is critical for cornea wound healing due to the controlled release rate and increased drug bioavailability. It should be mentioned that the transport of various types of medications into the different sections of the eye, particularly the cornea, is exceedingly challenging because of its distinctive structure and various barriers throughout the eye. Nanoparticles are being studied to improve medicine delivery strategies for ocular disease. Repetitive corneal drug delivery using biodegradable nanocarriers allows a medicine to remain in different parts of the cornea for extended periods of time and thus improve administration route effectiveness. In this review, we discussed eye anatomy, ocular delivery barriers, as well as the emphasis on the biodegradable nanomaterials ranging from organic nanostructures, such as nanomicelles, polymers, liposomes, niosomes, nanowafers, nanoemulsions, nanosuspensions, nanocrystals, cubosomes, olaminosomes, hybridized NPs, dendrimers, bilosomes, solid lipid NPs, nanostructured lipid carriers, and nanofiber to organic nanomaterials like silver, gold, and mesoporous silica nanoparticles. In addition, we describe the nanotechnology-based ophthalmic medications that are presently on the market or in clinical studies. Finally, drawing on current trends and therapeutic approaches, we discuss the challenges that innovative optical drug delivery systems confront and propose future research routes. We hope that this review will serve as a source of motivation and inspiration for developing innovative ophthalmic formulations.
Collapse
Affiliation(s)
- Milad Abbasi
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hossein Aghamollaei
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Ahmad Vaez
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Mohammad Amani
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hesam Kamyab
- Department of Biomaterials, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, 600077, India; The KU-KIST Graduate School of Energy and Environment, Korea University, 145 Anam-Ro, Seongbuk-Gu, Seoul, 02841, Republic of Korea; Universidad UTE, Quito, 170527, Ecuador.
| | - Shreeshivadasan Chelliapan
- Department of Smart Engineering and Advanced Technology, Faculty of Artificial Intelligence, Universiti Teknologi Malaysia, Jalan Sultan Yahya Petra, 54100, Kuala Lumpur, Malaysia.
| | - Sajad Jamalpour
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, 50603, Malaysia
| | - Renato Zambrano-Dávila
- Universidad UTE, Centro de Investigación en Salud Públicay Epidemiología Clínica (CISPEC), Quito, 170527, Ecuador
| |
Collapse
|
7
|
Su X, Wang H, Li Q, Chen Z. Extracellular Vesicles: A Review of Their Therapeutic Potentials, Sources, Biodistribution, and Administration Routes. Int J Nanomedicine 2025; 20:3175-3199. [PMID: 40098717 PMCID: PMC11913029 DOI: 10.2147/ijn.s502591] [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: 10/29/2024] [Accepted: 02/18/2025] [Indexed: 03/19/2025] Open
Abstract
Extracellular vesicles (EVs) participate in intercellular communication and play an essential role in physiological and pathological processes. In recent years, EVs have garnered significant attention as cell-free therapeutic alternatives, vectors for drug and gene delivery, biomarkers for disease diagnosis and prognosis, vaccine development, and nutraceuticals. The biodistribution of EVs critically influences their efficacy and toxicity. Therefore, this review aims to discuss the main factors influencing the biodistribution of unmodified EVs, highlighting their distribution patterns, advantages, limitations, and applications under different routes of administration. In addition, we provide a comprehensive discussion of the currently available sources of EVs and summarize the current status of the therapeutic potentials of EVs. By optimizing administration routes and selecting appropriate EV sources, we aim to offer valuable insights to enhance the delivery efficiency and therapeutic efficacy of EVs to target tissues.
Collapse
Affiliation(s)
- Xiaorong Su
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Hongxiang Wang
- Department of Hematology, Key Laboratory for Molecular Diagnosis of Hubei Province, the Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, People's Republic of China
| | - Qiubai Li
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
- Hubei Engineering Research Center for Application of Extracellular Vesicle, Hubei University of Science and Technology, Xianning, 437100, People's Republic of China
| | - Zhichao Chen
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| |
Collapse
|
8
|
Évora A, Garcia G, Rubi A, De Vitis E, Matos AT, Vaz AR, Gervaso F, Gigli G, Polini A, Brites D. Exosomes enriched with miR-124-3p show therapeutic potential in a new microfluidic triculture model that recapitulates neuron-glia crosstalk in Alzheimer's disease. Front Pharmacol 2025; 16:1474012. [PMID: 40144670 PMCID: PMC11936931 DOI: 10.3389/fphar.2025.1474012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 01/03/2025] [Indexed: 03/28/2025] Open
Abstract
Background Alzheimer's disease (AD), a complex neurodegenerative disease associated with ageing, is the leading cause of dementia. Few people with early AD are eligible for the novel Food and Drug Administration (FDA)-approved drug treatments. Accordingly, new tools and early diagnosis markers are required to predict subtypes, individual stages, and the most suitable personalized treatment. We previously demonstrated that the regulation of microRNA (miR)-124 is crucial for proper neuronal function and microglia reshaping in human AD cell models. Objective The aim of this study was to develop an efficient miR-124-3p-loaded exosome strategy and validate its therapeutic potential in using a multi-compartment microfluidic device of neuron-glia that recapitulates age-AD pathological features. Methods and results Using cortical microglia from mouse pups, separated from glial mixed cultures and maintained for 2 days in vitro (stressed microglia), we tested the effects of SH-SY5Y-derived exosomes loaded with miR-124-3p mimic either by their direct transfection with Exo-Fect™ (ET124) or by their isolation from the secretome of miR-124 transfected cells (CT124). ET124 revealed better delivery effciency and higher potent effects in improving the stressed microglia status than CT124. Tricultures of human SH-SY5Y neuroblastoma cells (SH-WT) were established in the presence of the human microglia cell line (HMC3) and immortalized human astrocytes (IM-HA) in tricompartmentalized microfluidic devices. Replacement of SH-WT cells with those transfected with APP695 (SH-SWE) in the tricultures and addition of low doses of hydrogen peroxide were used to simulate late-onset AD. The system mimicked AD-associated neurodegeneration and neuroinflammation processes. Notably, ET124 exhibited neuroprotective properties across the three cell types in the AD model by preventing neuronal apoptosis and neurite deficits, redirecting microglial profiles towards a steady state, and attenuating the inflammatory and miRNA fingerprints associated with astrocyte reactivity. Conclusion To the best of our knowledge, this is the first study supporting the neuro- and immunoprotective properties of miR-124-engineered exosomes in a microfluidic triculture platform, recapitulating age-related susceptibility to AD. Our system offers potential to develop personalized medicines in AD patient subtypes.
Collapse
Affiliation(s)
- Artemizia Évora
- Neuroinflammation, Signaling and Neuroregeneration, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Gonçalo Garcia
- Neuroinflammation, Signaling and Neuroregeneration, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Ana Rubi
- Neuroinflammation, Signaling and Neuroregeneration, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Eleonora De Vitis
- Institute of Nanotechnology, National Research Council (CNR Nanotec), Lecce, Italy
| | - Ana Teresa Matos
- Neuroinflammation, Signaling and Neuroregeneration, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Ana Rita Vaz
- Neuroinflammation, Signaling and Neuroregeneration, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Francesca Gervaso
- Institute of Nanotechnology, National Research Council (CNR Nanotec), Lecce, Italy
| | - Giuseppe Gigli
- Institute of Nanotechnology, National Research Council (CNR Nanotec), Lecce, Italy
- Dipartimento di Medicina Sperimentale, Università Del Salento, Lecce, Italy
| | - Alessandro Polini
- Institute of Nanotechnology, National Research Council (CNR Nanotec), Lecce, Italy
| | - Dora Brites
- Neuroinflammation, Signaling and Neuroregeneration, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
9
|
Li Y, Jiang J, Li J, Liu S, Wang C, Yu Z, Xia Y. Exosome-Derived CDC42 From Hypoxia-Pretreated Neural Stem Cells Inhibits ACSL4-Related Ferroptosis to Alleviate Vascular Injury in Parkinson's Disease Mice Models. J Neurochem 2025; 169:e70027. [PMID: 40035385 DOI: 10.1111/jnc.70027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 02/10/2025] [Accepted: 02/11/2025] [Indexed: 03/05/2025]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder that gets exacerbated by vascular injury. Neural stem cell-derived exosomes (NSC-Exos) display effective neuroprotective properties in PD models. Cell division control protein 42 (CDC42) is connected to angiogenesis, but its effects in PD remain undefined. This research aims to reveal the role of CDC42 in PD. First, we applied 1-methyl-4-phenylpyridinium (MPP+) to induce the human cerebral microvascular endothelial cells (HCMECs) model and evaluated cell viability and ferroptosis. Then, we characterized NSC-Exos. Next, to appraise the effect of hypoxia-pretreated NSC-Exos (H-NSC-Exos) on the MPP+-induced cells model, we examined angiogenesis and ferroptosis in HCMECs. Moreover, we constructed the PD mice model using 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and tested the behavioral experiments and vascular injury of mice. Furthermore, we examined cellular ferroptosis and angiogenesis after knockdown of CDC42. Additionally, we investigated the interaction of CDC42 with Acyl-CoA synthetase long-chain family member 4 (ACSL4) and detected cellular ferroptosis and angiogenesis after overexpression of ACSL4. We found that H-NSC-Exos reversed the MPP+-induced decrease in HCMECs viability and migration, lowered lipid-reactive oxygen species (lipid-ROS) levels, suppressed ferroptosis, and facilitated angiogenesis. Moreover, H-NSC-Exos attenuated MPTP-induced PD development, vascular injury, and ferroptosis in mice. H-NSC-Exos with the knockdown of CDC42 reduced cell viability and angiogenesis and raised ferroptosis and lipid-ROS levels, which were reversed by ferrostatin-1 and liproxstatin-1. CDC42 interacted with ACSL4. Furthermore, overexpression of ACSL4 aggravated the above effects of H-NSC-Exos in which CDC42 was knocked down. Our study reveals that H-NSC-Exos-derived CDC42 inhibited ACSL4-related ferroptosis to alleviate vascular injury in PD mice models. CDC42 may serve as a potent therapeutic target for PD treatment.
Collapse
Affiliation(s)
- You Li
- Department of Neurosurgery, Affiliated Haikou Hospital at Xiangya Medical College, Central South University, Haikou, China
| | - Junwen Jiang
- Department of Neurosurgery, Affiliated Haikou Hospital at Xiangya Medical College, Central South University, Haikou, China
| | - Jiameng Li
- Department of Neurosurgery, Affiliated Haikou Hospital at Xiangya Medical College, Central South University, Haikou, China
| | - Siliang Liu
- Department of Neurosurgery, Affiliated Haikou Hospital at Xiangya Medical College, Central South University, Haikou, China
| | - Chuang Wang
- Department of Neurosurgery, Affiliated Haikou Hospital at Xiangya Medical College, Central South University, Haikou, China
| | - Zhengtao Yu
- Department of Neurosurgery, Affiliated Haikou Hospital at Xiangya Medical College, Central South University, Haikou, China
| | - Ying Xia
- Department of Neurosurgery, Affiliated Haikou Hospital at Xiangya Medical College, Central South University, Haikou, China
| |
Collapse
|
10
|
Baghban R, Namvar E, Attar A, Mortazavi M. Progressing nanotechnology to improve diagnosis and targeted therapy of Diabetic Retinopathy. Biomed Pharmacother 2025; 183:117786. [PMID: 39753094 DOI: 10.1016/j.biopha.2024.117786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 02/08/2025] Open
Abstract
The inherent limitations of traditional treatments for Diabetic Retinopathy (DR) have spurred the development of various nanotechnologies, offering a safer and more efficient approach to managing the disease. Nanomedicine platforms present promising advancements in the diagnosis and treatment of DR by enhancing imaging capabilities, enabling targeted and controlled drug delivery. These innovations ultimately lead to more effective and personalized treatments with fewer side effects. This review highlights the progress, challenges, and opportunities in developing effective diagnostics and therapeutics for DR. Additionally, it explores innovative engineering techniques that leverage our growing understanding of nano-bio interactions to create more potent nanotherapeutics for patients.
Collapse
Affiliation(s)
- Roghayyeh Baghban
- Poostchi Ophthalmology Research Center, Department of Ophthalmology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ehsan Namvar
- Poostchi Ophthalmology Research Center, Department of Ophthalmology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Alireza Attar
- Poostchi Ophthalmology Research Center, Department of Ophthalmology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mojtaba Mortazavi
- Department of Biotechnology, Institute of Science and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran
| |
Collapse
|
11
|
Blanco-Agudín N, Ye S, González-Fernández S, Alcalde I, Merayo-Lloves J, Quirós LM. Exosomes in Ocular Health: Recent Insights into Pathology, Diagnostic Applications and Therapeutic Functions. Biomedicines 2025; 13:233. [PMID: 39857816 PMCID: PMC11762739 DOI: 10.3390/biomedicines13010233] [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: 11/28/2024] [Revised: 01/09/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025] Open
Abstract
Exosomes are extracellular vesicles ranging from 30 to 150 nm in diameter that contain proteins, nucleic acids and other molecules. Produced by virtually all cell types, they travel throughout the body until they reach their target, where they can trigger a wide variety of effects by transferring the molecular cargo to recipient cells. In the context of ocular physiology, exosomes play a very important role in embryological development, the regulation of homeostasis and the immune system, which is crucial for normal vision. Consequently, in pathological situations, exosomes also undergo modifications in terms of quantity, composition and content, depending on the etiology of the disease. However, the mechanisms by which exosomes contribute to ocular pathology has not yet been studied in depth, and many questions remain unanswered. This review aims to summarize the most recent knowledge on the function of exosomes in the ocular system in healthy individuals and the role they play during pathological processes of a degenerative, infectious, neurodegenerative, vascular and inflammatory nature, such as keratoconus, keratitis, glaucoma, diabetic retinopathy and uveitis. Furthermore, given their unique characteristics, their potential as diagnostic biomarkers or therapeutic agents and their application in clinical ophthalmology are also explored, along with the main limitations that researchers face today in the field.
Collapse
Affiliation(s)
- Noelia Blanco-Agudín
- Department of Functional Biology, University of Oviedo, 33006 Oviedo, Spain; (N.B.-A.); (S.Y.); (S.G.-F.)
- Instituto Universitario Fernández-Vega, Fundación de Investigación Oftalmológica, University of Oviedo, 33012 Oviedo, Spain;
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Suhui Ye
- Department of Functional Biology, University of Oviedo, 33006 Oviedo, Spain; (N.B.-A.); (S.Y.); (S.G.-F.)
- Instituto Universitario Fernández-Vega, Fundación de Investigación Oftalmológica, University of Oviedo, 33012 Oviedo, Spain;
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Sara González-Fernández
- Department of Functional Biology, University of Oviedo, 33006 Oviedo, Spain; (N.B.-A.); (S.Y.); (S.G.-F.)
- Instituto Universitario Fernández-Vega, Fundación de Investigación Oftalmológica, University of Oviedo, 33012 Oviedo, Spain;
| | - Ignacio Alcalde
- Instituto Universitario Fernández-Vega, Fundación de Investigación Oftalmológica, University of Oviedo, 33012 Oviedo, Spain;
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Jesús Merayo-Lloves
- Instituto Universitario Fernández-Vega, Fundación de Investigación Oftalmológica, University of Oviedo, 33012 Oviedo, Spain;
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Luis M. Quirós
- Department of Functional Biology, University of Oviedo, 33006 Oviedo, Spain; (N.B.-A.); (S.Y.); (S.G.-F.)
- Instituto Universitario Fernández-Vega, Fundación de Investigación Oftalmológica, University of Oviedo, 33012 Oviedo, Spain;
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
| |
Collapse
|
12
|
Xu K, Feng H, Zhao R, Huang Y. Targeting Tetraspanins at Cell Interfaces: Functional Modulation and Exosome-Based Drug Delivery for Precise Disease Treatment. ChemMedChem 2025; 20:e202400664. [PMID: 39415492 DOI: 10.1002/cmdc.202400664] [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: 08/26/2024] [Revised: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 10/18/2024]
Abstract
Tetraspanins are key players in various physiological and pathological processes, including malignancy, immune response, fertilization, and infectious disease. Affinity ligands targeting the interactions between tetraspanins and partner proteins are promising for modulating downstream signaling pathways, thus emerging as attractive candidates for interfering related biological functions. Due to the involvement in vesicle biogenesis and cargo trafficking, tetraspanins are also regarded as exosome markers, and become molecular targets for drug loading and delivery. Given the rapid development in these areas, this minireview focuses on recent advances in design and engineering of affinity binders toward tetraspanins including CD63, CD81, and CD9. Their mechanism of actions in modulating protein interactions at cell interfaces and treatment of malignant diseases are discussed. Strategies for constructing exosome-based drug delivery platforms are also reviewed, with emphasis on the important roles of tetraspanins and the affinity ligands. Finally, challenges and future development of tetraspanin-targeting therapy and exosomal drug delivery platforms are also discussed.
Collapse
Affiliation(s)
- Kun Xu
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Huixia Feng
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Rui Zhao
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yanyan Huang
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
13
|
Sun H, Zou Y, Chen Z, He Y, Ye K, Liu H, Qiu L, Zhang Y, Mai Y, Chen X, Mao Z, Wang W, Yi C. Nanodrug-Engineered Exosomes Achieve a Jointly Dual-Pathway Inhibition on Cuproptosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413408. [PMID: 39639737 PMCID: PMC11775538 DOI: 10.1002/advs.202413408] [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: 10/22/2024] [Revised: 11/18/2024] [Indexed: 12/07/2024]
Abstract
Cuproptosis, caused by an intracellular overload of copper (Cu) ions and overexpression of ferredoxin 1 (FDX1), is identified for its regulatory role in the skin wound healing process. This study verifies the presence of cuproptosis in skin wound beds and reactive oxygen species-induced cells model. To address the two pathways leading to cell cuproptosis, a nanodrug-engineered exosomes is proposed. A Cu-chelator (Clioquinol, CQ) polydopamine (PDA)-modified stem cell exosome loaded with siRNA-FDX1, named EXOsiFDX1-PDA@CQ, is designed to efficiently inhibit the two cuproptosis pathways. The functionalized exosomes are loaded into an injectable hydrogel and applied to treat diabetic wounds in mice and acute skin wounds in pigs. The local and controlled release of EXOsiFDX1-PDA@CQ ensures the retention of the therapeutic agent at wound beds, effectively promoting wound healing. The strategy of engineered exosomes with functional nanoparticles (NPs) proposed in this study offers an efficient and scalable new approach for regulating cuproptosis.
Collapse
Affiliation(s)
- Hanxiao Sun
- The Second Affiliated Hospital of Zhejiang University College of MedicineHangzhou310000China
| | - Yang Zou
- College of Chemical and Biological EngineeringZhejiang UniversityHangzhouZhejiang310027China
| | - Zhengtai Chen
- The Second Affiliated Hospital of Zhejiang University College of MedicineHangzhou310000China
| | - Yan He
- The Second Affiliated Hospital of Zhejiang University College of MedicineHangzhou310000China
| | - Kai Ye
- The Second Affiliated Hospital of Zhejiang University College of MedicineHangzhou310000China
| | - Huan Liu
- The Second Affiliated Hospital of Zhejiang University College of MedicineHangzhou310000China
| | - Lihong Qiu
- The Second Affiliated Hospital of Zhejiang University College of MedicineHangzhou310000China
| | - Yufan Zhang
- The Second Affiliated Hospital of Zhejiang University College of MedicineHangzhou310000China
| | - Yuexue Mai
- The Second Affiliated Hospital of Zhejiang University College of MedicineHangzhou310000China
| | - Xinghong Chen
- The Second Affiliated Hospital of Zhejiang University College of MedicineHangzhou310000China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationDepartment of Polymer Science and EngineeringZhejiang UniversityHangzhouZhejiang310027China
| | - Wei Wang
- The Second Affiliated Hospital of Zhejiang University College of MedicineHangzhou310000China
- College of Chemical and Biological EngineeringZhejiang UniversityHangzhouZhejiang310027China
| | - Chenggang Yi
- The Second Affiliated Hospital of Zhejiang University College of MedicineHangzhou310000China
| |
Collapse
|
14
|
Lei Y, Liu Q, Chen B, Wu F, Li Y, Dong X, Ma N, Wu Z, Zhu Y, Wang L, Fu Y, Liu Y, Song Y, Du M, Zhang H, Zhu J, Lyons TJ, Wang T, Hu J, Xu H, Chen M, Yan H, Wang X. Protein O-GlcNAcylation coupled to Hippo signaling drives vascular dysfunction in diabetic retinopathy. Nat Commun 2024; 15:9334. [PMID: 39472558 PMCID: PMC11522279 DOI: 10.1038/s41467-024-53601-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/17/2024] [Indexed: 11/02/2024] Open
Abstract
Metabolic disorder significantly contributes to diabetic vascular complications, including diabetic retinopathy, the leading cause of blindness in the working-age population. However, the molecular mechanisms by which disturbed metabolic homeostasis causes vascular dysfunction in diabetic retinopathy remain unclear. O-GlcNAcylation modification acts as a nutrient sensor particularly sensitive to ambient glucose. Here, we observe pronounced O-GlcNAc elevation in retina endothelial cells of diabetic retinopathy patients and mouse models. Endothelial-specific depletion or pharmacological inhibition of O-GlcNAc transferase effectively mitigates vascular dysfunction. Mechanistically, we find that Yes-associated protein (YAP) and Transcriptional co-activator with PDZ-binding motif (TAZ), key effectors of the Hippo pathway, are O-GlcNAcylated in diabetic retinopathy. We identify threonine 383 as an O-GlcNAc site on YAP, which inhibits its phosphorylation at serine 397, leading to its stabilization and activation, thereby promoting vascular dysfunction by inducing a pro-angiogenic and glucose metabolic transcriptional program. This work emphasizes the critical role of the O-GlcNAc-Hippo axis in the pathogenesis of diabetic retinopathy and suggests its potential as a therapeutic target.
Collapse
Affiliation(s)
- Yi Lei
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, Tianjin, China
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Qiangyun Liu
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, Tianjin, China
| | - Binggui Chen
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Fangfang Wu
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yiming Li
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, Tianjin, China
| | - Xue Dong
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, Tianjin, China
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Nina Ma
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Ziru Wu
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yanfang Zhu
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, Tianjin, China
| | - Lu Wang
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yuxin Fu
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yuming Liu
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, Tianjin, China
| | - Yinting Song
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, Tianjin, China
| | - Mei Du
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, Tianjin, China
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Heng Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Jidong Zhu
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Timothy J Lyons
- Division of Endocrinology, Diabetes and Metabolic Diseases at the Medical University of South Carolina, Charleston, SC, USA
| | - Ting Wang
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Junhao Hu
- Laboratory of Vascular Biology and Organ Homeostasis, Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Heping Xu
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Mei Chen
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Hua Yan
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, Tianjin, China.
- School of Medicine, Nankai University, Tianjin, China.
| | - Xiaohong Wang
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, Tianjin, China.
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
15
|
Chen S, Liu Y, Zhang Y, Guo X, Bai T, He K, Zhu Y, Lei Y, Du M, Wang X, Liu Q, Yan H. Bruton's tyrosine kinase inhibition suppresses pathological retinal angiogenesis. Br J Pharmacol 2024. [PMID: 39374939 DOI: 10.1111/bph.17344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 06/25/2024] [Accepted: 08/22/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND AND PURPOSE Pathological retinal angiogenesis is a typical manifestation of vision-threatening ocular diseases. Many patients exhibit poor response or resistance to anti-vascular endothelial growth factor (VEGF) agents. Bruton's tyrosine kinase (BTK) controls the proliferation and function of immune cells. Therefore, we examined the anti-inflammatory and anti-angiogenic effects of BTK inhibition on retinal angiogenesis. EXPERIMENTAL APPROACH Retinal neovascularisation and vascular leakage in oxygen-induced retinopathy in C57/BL6J mice were assessed by whole-mount retinal immunofluorescence. PLX5622 was used to deplete microglia and Rag1-knockout mice were used to test the contribution of lymphocytes to the effects of BTK inhibition. The cytokines, activation markers, inflammatory and immune-regulatory activities of retinal microglia/macrophages were detected using qRT-PCR and immunofluorescence. NLRP3 was detected by western blotting, and the effects of BTK inhibition on the co-culture of microglia and human retinal microvascular endothelial cells (HRMECs) were examined. KEY RESULTS BTK inhibition suppressed pathological angiogenesis and vascular leakage, and significantly reduced retinal inflammation, which involved microglia/macrophages but not lymphocytes. BTK inhibition increased anti-inflammatory factors and reduced pro-inflammatory cytokines that resulted from NLRP3 inflammasome activation. BTK inhibition suppressed the inflammatory activity of microglia/macrophages, and acted synergistically with anti-VEGF without retinal toxicity. Moreover, the supernatant of microglia incubated with BTK-inhibitor reduced the proliferation, tube formation and sprouting of HRMECs. CONCLUSION AND IMPLICATIONS BTK inhibition suppressed retinal neovascularisation and vascular leakage by modulating the inflammatory activity of microglia and macrophages. Our study suggests BTK inhibition as a novel and promising approach for alleviating pathological retinal angiogenesis.
Collapse
Affiliation(s)
- Siyue Chen
- Department of Ophthalmology, Tianjin Medical University General Hospital, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Key Laboratory of Ocular Trauma, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin, China
| | - Yuming Liu
- Department of Ophthalmology, Tianjin Medical University General Hospital, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Key Laboratory of Ocular Trauma, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin, China
| | - Yutian Zhang
- Department of Ophthalmology, Tianjin Medical University General Hospital, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Key Laboratory of Ocular Trauma, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin, China
| | - Xu Guo
- Department of Ophthalmology, Tianjin Medical University General Hospital, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Key Laboratory of Ocular Trauma, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin, China
| | - Tinghui Bai
- Department of Ophthalmology, Tianjin Medical University General Hospital, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Key Laboratory of Ocular Trauma, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin, China
| | - Kai He
- Department of Ophthalmology, Tianjin Medical University General Hospital, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Key Laboratory of Ocular Trauma, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin, China
| | - Yanfang Zhu
- Department of Ophthalmology, Tianjin Medical University General Hospital, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Key Laboratory of Ocular Trauma, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin, China
| | - Yi Lei
- Department of Ophthalmology, Tianjin Medical University General Hospital, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Key Laboratory of Ocular Trauma, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin, China
| | - Mei Du
- Department of Ophthalmology, Tianjin Medical University General Hospital, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Key Laboratory of Ocular Trauma, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin, China
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xiaohong Wang
- Department of Ophthalmology, Tianjin Medical University General Hospital, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Key Laboratory of Ocular Trauma, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin, China
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Qiang Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Hua Yan
- Department of Ophthalmology, Tianjin Medical University General Hospital, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Key Laboratory of Ocular Trauma, Tianjin Institute of Eye Health and Eye Diseases, China-UK "Belt and Road" Ophthalmology Joint Laboratory, Tianjin, China
| |
Collapse
|
16
|
Su Y, Chen M, Xu W, Gu P, Fan X. Advances in Extracellular-Vesicles-Based Diagnostic and Therapeutic Approaches for Ocular Diseases. ACS NANO 2024; 18:22793-22828. [PMID: 39141830 PMCID: PMC11363148 DOI: 10.1021/acsnano.4c08486] [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: 06/25/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/16/2024]
Abstract
Extracellular vesicles (EVs) are nanoscale membrane vesicles of various sizes that can be secreted by most cells. EVs contain a diverse array of cargo, including RNAs, lipids, proteins, and other molecules with functions of intercellular communication, immune modulation, and regulation of physiological and pathological processes. The biofluids in the eye, including tears, aqueous humor, and vitreous humor, are important sources for EV-based diagnosis of ocular disease. Because the molecular cargos may reflect the biology of their parental cells, EVs in these biofluids, as well as in the blood, have been recognized as promising candidates as biomarkers for early diagnosis of ocular disease. Moreover, EVs have also been used as therapeutics and targeted drug delivery nanocarriers in many ocular disorders because of their low immunogenicity and superior biocompatibility in nature. In this review, we provide an overview of the recent advances in the field of EV-based studies on the diagnosis and therapeutics of ocular disease. We summarized the origins of EVs applied in ocular disease, assessed different methods for EV isolation from ocular biofluid samples, highlighted bioengineering strategies of EVs as drug delivery systems, introduced the latest applications in the diagnosis and treatment of ocular disease, and presented their potential in the current clinical trials. Finally, we briefly discussed the challenges of EV-based studies in ocular disease and some issues of concern for better focusing on clinical translational studies of EVs in the future.
Collapse
Affiliation(s)
- Yun Su
- Department
of Ophthalmology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- Shanghai
Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| | - Moxin Chen
- Department
of Ophthalmology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- Shanghai
Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| | - Wei Xu
- Department
of Ophthalmology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- Shanghai
Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| | - Ping Gu
- Department
of Ophthalmology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- Shanghai
Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| | - Xianqun Fan
- Department
of Ophthalmology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- Shanghai
Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, China
| |
Collapse
|
17
|
Yu F, Zhao X, Wang Q, Fang PH, Liu L, Du X, Li W, He D, Zhang T, Bai Y, Liu L, Li S, Yuan J. Engineered Mesenchymal Stromal Cell Exosomes-Loaded Microneedles Improve Corneal Healing after Chemical Injury. ACS NANO 2024. [PMID: 39047084 DOI: 10.1021/acsnano.4c00423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
Corneal alkali burns represent a prevalent ophthalmic emergency with the potential to induce blindness. The main contributing mechanisms include excessive inflammation and delayed wound healing. Existing clinical therapies have limitations, promoting the exploration of alternative methods that offer improved efficacy and reduced side effects. Adipose-derived stem cell-exosome (ADSC-Exo) has the potential to sustain immune homeostasis and facilitate tissue regeneration. Nevertheless, natural ADSC-Exo lacks disease specificity and exhibits limited bioavailability on the ocular surface. In this study, we conjugated antitumor necrosis factor-α antibodies (aT) to the surface of ADSC-Exo using matrix metalloproteinase-cleavable peptide chains to create engineered aT-Exo with synergistic effects. In both in vivo and in vitro assessments, aT-Exo demonstrated superior efficacy in mitigating corneal injuries compared to aT alone, unmodified exosomes, or aT simply mixed with exosomes. The cleavable conjugation of aT-Exo notably enhanced wound healing and alleviated inflammation more effectively. Simultaneously, we developed poly(vinyl alcohol) microneedles (MNs) for precise and sustained exosome delivery. The in vivo results showcased the superior therapeutic efficiency of MNs compared with conventional topical administration and subconjunctival injection. Therefore, the bioactive nanodrugs-loaded MNs treatment presents a promising strategy for addressing ocular surface diseases.
Collapse
Affiliation(s)
- Fei Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510623, China
| | - Xuan Zhao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510623, China
| | - Qian Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510623, China
| | - Po-Han Fang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510623, China
| | - Liu Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510623, China
| | - Xinyue Du
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510623, China
| | - Weihua Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510623, China
| | - Dalian He
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510623, China
| | - Tingting Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510623, China
| | - Ying Bai
- Guangdong Engineering Technology Research Centre for Functional Biomaterials, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510006, China
| | - Lu Liu
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR 999077, China
| | - Saiqun Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510623, China
| | - Jin Yuan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510623, China
| |
Collapse
|
18
|
Li B, Zhang M, Chen S, Zhao C, Li X, Zhang X. Small extracellular vesicle-based delivery of interleukin-10 improves treatment of experimental autoimmune uveitis. Exp Eye Res 2024; 244:109936. [PMID: 38763351 DOI: 10.1016/j.exer.2024.109936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/05/2024] [Accepted: 05/17/2024] [Indexed: 05/21/2024]
Abstract
Non-infectious uveitis is an intraocular autoimmune disease mainly characterized by immune dysregulation of the eye, which may cause blindness if not well treated. Interleukin 10 (IL-10) is a potent cytokine with multiple immunoregulatory functions. However, it's in vivo activity is unstable owing to its inherent protein instability and short plasma half-life. Therefore, our previous research tried to establish IL-10-overexpressing MSC-sEVs (sEVs-IL10) using lentiviral transfection. While this approach indeed improved drug delivery, it also suffered from tedious procedures and limited loading efficiency. Accordingly, we constructed a novel MSC-sEVs-based delivery system for IL-10 (IL-10@sEVs) by sonication. The obtained formulation (IL-10@sEVs) had relatively higher loading efficiency and exerted a more profound immunomodulatory effect than sEVs-IL10 in vitro. Furthermore, IL-10@sEVs had significant therapeutic effects in a mouse model of experimental autoimmune uveitis (EAU) by decreasing the percentage of Th17 cells, increasing regulatory T cells in the eye, and draining lymph nodes. In summary, sonication outperforms conventional transfection methods for loading IL-10 into MSC-sEVs.
Collapse
Affiliation(s)
- Baiyi Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China
| | - Mi Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China
| | - Shuang Chen
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China
| | - Chuan Zhao
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China
| | - Xiaorong Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China
| | - Xiaomin Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China.
| |
Collapse
|
19
|
Liu X, Huang K, Zhang F, Huang G, Wang L, Wu G, Ren H, Yang G, Lin Z. Multifunctional nano-in-micro delivery systems for targeted therapy in fundus neovascularization diseases. J Nanobiotechnology 2024; 22:354. [PMID: 38902775 PMCID: PMC11191225 DOI: 10.1186/s12951-024-02614-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 06/03/2024] [Indexed: 06/22/2024] Open
Abstract
Fundus neovascularization diseases are a series of blinding eye diseases that seriously impair vision worldwide. Currently, the means of treating these diseases in clinical practice are continuously evolving and have rapidly revolutionized treatment opinions. However, key issues such as inadequate treatment effectiveness, high rates of recurrence, and poor patient compliance still need to be urgently addressed. Multifunctional nanomedicine can specifically respond to both endogenous and exogenous microenvironments, effectively deliver drugs to specific targets and participate in activities such as biological imaging and the detection of small molecules. Nano-in-micro (NIM) delivery systems such as metal, metal oxide and up-conversion nanoparticles (NPs), quantum dots, and carbon materials, have shown certain advantages in overcoming the presence of physiological barriers within the eyeball and are widely used in the treatment of ophthalmic diseases. Few studies, however, have evaluated the efficacy of NIM delivery systems in treating fundus neovascular diseases (FNDs). The present study describes the main clinical treatment strategies and the adverse events associated with the treatment of FNDs with NIM delivery systems and summarizes the anatomical obstacles that must be overcome. In this review, we wish to highlight the principle of intraocular microenvironment normalization, aiming to provide a more rational approach for designing new NIM delivery systems to treat specific FNDs.
Collapse
Affiliation(s)
- Xin Liu
- Department of Ophthalmology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Keke Huang
- Department of Ophthalmology, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China
| | - Fuxiao Zhang
- Department of Ophthalmology, The Second People's Hospital of Chengdu, The Affiliated Hospital of Chengdu Medical College, Chengdu, 610031, China
| | - Ge Huang
- Department of Ophthalmology, The Second People's Hospital of Chengdu, The Affiliated Hospital of Chengdu Medical College, Chengdu, 610031, China
| | - Lu Wang
- Department of Ophthalmology, The Second People's Hospital of Chengdu, The Affiliated Hospital of Chengdu Medical College, Chengdu, 610031, China
| | - Guiyu Wu
- Department of Ophthalmology, The Second People's Hospital of Chengdu, The Affiliated Hospital of Chengdu Medical College, Chengdu, 610031, China
| | - Hui Ren
- Department of Ophthalmology, The Second People's Hospital of Chengdu, The Affiliated Hospital of Chengdu Medical College, Chengdu, 610031, China.
| | - Guang Yang
- Department of Ophthalmology, The Second People's Hospital of Chengdu, The Affiliated Hospital of Chengdu Medical College, Chengdu, 610031, China.
| | - Zhiqing Lin
- Department of Ophthalmology, The Second People's Hospital of Chengdu, The Affiliated Hospital of Chengdu Medical College, Chengdu, 610031, China.
| |
Collapse
|
20
|
Peng X, Zhang T, Liu R, Jin X. Potential in exosome-based targeted nano-drugs and delivery vehicles for posterior ocular disease treatment: from barriers to therapeutic application. Mol Cell Biochem 2024; 479:1319-1333. [PMID: 37402019 DOI: 10.1007/s11010-023-04798-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/21/2023] [Indexed: 07/05/2023]
Abstract
Posterior ocular disease, a disease that accounts for 55% of all ocular diseases, can contribute to permanent vision loss if left without treatment. Due to the special structure of the eye, various obstacles make it difficult for drugs to reach lesions in the posterior ocular segment. Therefore, the development of highly permeable targeted drugs and delivery systems is particularly important. Exosomes are a class of extracellular vesicles at 30-150 nm, which are secreted by various cells, tissues, and body fluids. They carry various signaling molecules, thus endowing them with certain physiological functions. In this review, we describe the ocular barriers and the biogenesis, isolation, and engineering of exosomes, as exosomes not only have pharmacological effects but also are good nanocarriers with targeted properties. Moreover, their biocompatibility and immunogenicity are better than synthetic nanocarriers. Most importantly, they may have the ability to pass through the blood-eye barrier. Thus, they may be developed as both targeted nano-drugs and nano-delivery vehicles for the treatment of posterior ocular diseases. We focus on the current status and potential application of exosomes as targeted nano-drugs and nano-delivery vehicles in posterior ocular diseases.
Collapse
Affiliation(s)
- Xingru Peng
- State Key Laboratory of Component‑based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Tingting Zhang
- State Key Laboratory of Component‑based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Rui Liu
- State Key Laboratory of Component‑based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Xin Jin
- Department of Health Services, Logistics University of People's Armed Police Force, Tianjin, Chenlin Road, Hedong District, Tianjin, 300162, China.
| |
Collapse
|
21
|
Liu S, Yan Z, Huang Z, Yang H, Li J. Smart Nanocarriers for the Treatment of Retinal Diseases. ACS APPLIED BIO MATERIALS 2024; 7:2070-2085. [PMID: 38489843 DOI: 10.1021/acsabm.3c01289] [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: 03/17/2024]
Abstract
Retinal diseases, such as age-related macular degeneration, diabetic retinopathy, and retinoblastoma, stand as the leading causes of irreversible vision impairment and blindness worldwide. Effectively administering drugs for retinal diseases poses a formidable challenge due to the presence of complex ocular barriers and elimination mechanisms. Over time, various approaches have been developed to fabricate drug delivery systems for improving retinal therapy including virus vectors, lipid nanoparticles, and polymers. However, conventional nanocarriers encounter issues related to the controllability, efficiency, and safety in the retina. Therefore, the development of smart nanocarriers for effective or more invasive long-term treatment remains a desirable goal. Recently, approaches have surfaced for the intelligent design of nanocarriers, leveraging specific responses to external or internal triggers and enabling multiple functions for retinal therapy such as topical administration, prolonged drug release, and site-specific drug delivery. This Review provides an overview of prevalent retinal pathologies and related pharmacotherapies to enhance the understanding of retinal diseases. It also surveys recent developments and strategies employed in the intelligent design of nanocarriers for retinal disease. Finally, the challenges of smart nanocarriers in potential clinical retinal therapeutic applications are discussed to inspire the next generation of smart nanocarriers.
Collapse
Affiliation(s)
- Shuya Liu
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University, Fuzhou 350116, P. R. China
| | - Zhike Yan
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University, Fuzhou 350116, P. R. China
| | - Zixiang Huang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University, Fuzhou 350116, P. R. China
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University, Fuzhou 350116, P. R. China
| | - Jingying Li
- College of Biological Science and Engineering, Fuzhou University, Fuzhou 350116, P. R. China
| |
Collapse
|
22
|
Wu Y, Wang J, Pan T, Lei J, Fan Y, Wang J, Xu C, Gu Q, Wang X, Xiao T, Liu Q, Xie P, Hu Z. Human lens epithelial-secreted exosomes attenuate ocular angiogenesis via inhibiting microglial activation. Exp Eye Res 2024; 241:109837. [PMID: 38382576 DOI: 10.1016/j.exer.2024.109837] [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: 09/04/2023] [Revised: 02/05/2024] [Accepted: 02/18/2024] [Indexed: 02/23/2024]
Abstract
The lens is an avascular tissue, where epithelial cells (LECs) are the primary living cells. The role of LECs-derived exosomes (LEC-exos) is largely unknown. In our study, we determined the anti-angiogenic role of LEC-exos, manifested as regressed retinal neovascularization (NV) using the oxygen-induced retinopathy (OIR), and reduced choroidal NV size and pathological vascular leakage using the laser-induced choroidal neovascularization (laser-induced CNV). Furthermore, the activation and accumulation of microglia were also restricted by LEC-exos. Based on Luminex multiplex assays, the expressions of chemokines such as SCYB16/CXCL16, MCP-1/CCL2, I-TAC/CXCL11, and MIP 3beta/CCL19 were decreased after treatment with LEC-exos. Transwell assays showed that LEC-exos restricted the migration of the mouse microglia cell line (BV2 cells). After incubation with LEC-exos-treated BV2 cells, human umbilical vein endothelial cells (hUVECs) were collected for further evaluation using tube formation, Transwell assays, and 5-ethynyl-2'-deoxyuridine (EDU) assays. Using in vitro experiments, the pro-angiogenic effect of microglia was restricted by LEC-exos. Hence, it was investigated that LEC-exos attenuated ocular NV, which might attribute to the inhibition of microglial activation and accumulation.
Collapse
Affiliation(s)
- Yan Wu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Jiagui Wang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China; Department of Ophthalmology, Luhe People's Hospital of Nanjing, Nanjing, 211599, Jiangsu, China
| | - Ting Pan
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China; Department of Ophthalmology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Jie Lei
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Yuanyuan Fan
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Jingfan Wang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Changlin Xu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Qinyuan Gu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Xingxing Wang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Tianhao Xiao
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Qinghuai Liu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Ping Xie
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| | - Zizhong Hu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
23
|
Han G, Zhang Y, Zhong L, Wang B, Qiu S, Song J, Lin C, Zou F, Wu J, Yu H, Liang C, Wen K, Seow Y, Yin H. Generalizable anchor aptamer strategy for loading nucleic acid therapeutics on exosomes. EMBO Mol Med 2024; 16:1027-1045. [PMID: 38448545 PMCID: PMC11018858 DOI: 10.1038/s44321-024-00049-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/15/2024] [Accepted: 02/19/2024] [Indexed: 03/08/2024] Open
Abstract
Clinical deployment of oligonucleotides requires delivery technologies that improve stability, target tissue accumulation and cellular internalization. Exosomes show potential as ideal delivery vehicles. However, an affordable generalizable system for efficient loading of oligonucleotides on exosomes remain lacking. Here, we identified an Exosomal Anchor DNA Aptamer (EAA) via SELEX against exosomes immobilized with our proprietary CP05 peptides. EAA shows high binding affinity to different exosomes and enables efficient loading of nucleic acid drugs on exosomes. Serum stability of thrombin inhibitor NU172 was prolonged by exosome-loading, resulting in increased blood flow after injury in vivo. Importantly, Duchenne Muscular Dystrophy PMO can be readily loaded on exosomes via EAA (EXOEAA-PMO). EXOEAA-PMO elicited significantly greater muscle cell uptake, tissue accumulation and dystrophin expression than PMO in vitro and in vivo. Systemic administration of EXOEAA-PMO elicited therapeutic levels of dystrophin restoration and functional improvements in mdx mice. Altogether, our study demonstrates that EAA enables efficient loading of different nucleic acid drugs on exosomes, thus providing an easy and generalizable strategy for loading nucleic acid therapeutics on exosomes.
Collapse
Affiliation(s)
- Gang Han
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics & International Joint Laboratory of Ocular Diseases (Ministry of Education), School of Medical Technology & School of Basic Medical Sciences, Tianjin Medical University, Qixiangtai Road, Heping District, 300070, Tianjin, China
| | - Yao Zhang
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics & International Joint Laboratory of Ocular Diseases (Ministry of Education), School of Medical Technology & School of Basic Medical Sciences, Tianjin Medical University, Qixiangtai Road, Heping District, 300070, Tianjin, China
| | - Li Zhong
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics & International Joint Laboratory of Ocular Diseases (Ministry of Education), School of Medical Technology & School of Basic Medical Sciences, Tianjin Medical University, Qixiangtai Road, Heping District, 300070, Tianjin, China
| | - Biaobiao Wang
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics & International Joint Laboratory of Ocular Diseases (Ministry of Education), School of Medical Technology & School of Basic Medical Sciences, Tianjin Medical University, Qixiangtai Road, Heping District, 300070, Tianjin, China
| | - Shuai Qiu
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics & International Joint Laboratory of Ocular Diseases (Ministry of Education), School of Medical Technology & School of Basic Medical Sciences, Tianjin Medical University, Qixiangtai Road, Heping District, 300070, Tianjin, China
| | - Jun Song
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics & International Joint Laboratory of Ocular Diseases (Ministry of Education), School of Medical Technology & School of Basic Medical Sciences, Tianjin Medical University, Qixiangtai Road, Heping District, 300070, Tianjin, China
| | - Caorui Lin
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics & International Joint Laboratory of Ocular Diseases (Ministry of Education), School of Medical Technology & School of Basic Medical Sciences, Tianjin Medical University, Qixiangtai Road, Heping District, 300070, Tianjin, China
| | - Fangdi Zou
- Public Laboratory & Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center & Tianjin's Clinical Research Center for Cancer, 300060, Tianjin, China
| | - Jingqiao Wu
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics & International Joint Laboratory of Ocular Diseases (Ministry of Education), School of Medical Technology & School of Basic Medical Sciences, Tianjin Medical University, Qixiangtai Road, Heping District, 300070, Tianjin, China
| | - Huanan Yu
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics & International Joint Laboratory of Ocular Diseases (Ministry of Education), School of Medical Technology & School of Basic Medical Sciences, Tianjin Medical University, Qixiangtai Road, Heping District, 300070, Tianjin, China
| | - Chao Liang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, China
| | - Ke Wen
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, School of Basic Medical Sciences, Tianjin Medical University, 300070, Tianjin, China
| | - Yiqi Seow
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis St, Genome, Singapore, 138672, Republic of Singapore
| | - HaiFang Yin
- State Key Laboratory of Experimental Hematology & The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics & International Joint Laboratory of Ocular Diseases (Ministry of Education), School of Medical Technology & School of Basic Medical Sciences, Tianjin Medical University, Qixiangtai Road, Heping District, 300070, Tianjin, China.
- Department of Clinical Laboratory, Tianjin Medical University General Hospital, 300052, Tianjin, China.
| |
Collapse
|
24
|
Haroon K, Zheng H, Wu S, Liu Z, Tang Y, Yang GY, Liu Y, Zhang Z. Engineered exosomes mediated targeted delivery of neuroprotective peptide NR2B9c for the treatment of traumatic brain injury. Int J Pharm 2024; 649:123656. [PMID: 38040392 DOI: 10.1016/j.ijpharm.2023.123656] [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: 09/12/2023] [Revised: 11/19/2023] [Accepted: 11/28/2023] [Indexed: 12/03/2023]
Abstract
Neuroprotection is one of the core treatment strategies for brain injuries including traumatic brain injury (TBI). NR2B9c is a promising neuroprotective peptide but its clinical translation is limited because of poor brain penetrability. Exosomes are naturally occurring nanovesicles having therapeutic potential for TBI as well as an efficient drug delivery carrier to the brain. Here, we engineered exosomes with neuron targeting peptide rabies virus glycoprotein (RVG29) via bio-orthogonal click chemistry technique and loaded it with NR2B9c, developing RVG-ExoNR2B9c. RVG29 conjugated exosome had higher neuron targeting efficiency compared to naïve exosomes both in vivo and in vitro. RVG-ExoNR2B9c had great cytoprotective effect against oxygen glucose deprived Neuro2a cells. Intravenous administration of RVG-ExoNR2B9c significantly improved behavioral outcomes and reduced the lesion volume after TBI injury in a mice controlled cortical impact model. Due to their multifunctionality and significant efficacy, we anticipate that RVG-ExoNR2B9c have the potential to be translated both as therapeutic agent as well as cargo delivery system to the brain for the treatment of TBI.
Collapse
Affiliation(s)
- Khan Haroon
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Haoran Zheng
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Shengju Wu
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Ze Liu
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yaohui Tang
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Guo-Yuan Yang
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China.
| | - Yingli Liu
- Department of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University, School of Medicine, 639 Zhizaoju Road, Shanghai 200025, China.
| | - Zhijun Zhang
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China.
| |
Collapse
|
25
|
Manai F, Smedowski A, Kaarniranta K, Comincini S, Amadio M. Extracellular vesicles in degenerative retinal diseases: A new therapeutic paradigm. J Control Release 2024; 365:448-468. [PMID: 38013069 DOI: 10.1016/j.jconrel.2023.11.035] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 11/03/2023] [Accepted: 11/18/2023] [Indexed: 11/29/2023]
Abstract
Nanoscale extracellular vesicles (EVs), consisting of exomers, exosomes and microvesicles/ectosomes, have been extensively investigated in the last 20 years, although their biological role is still something of a mystery. EVs are involved in the transfer of lipids, nucleic acids and proteins from donor to recipient cells or distant organs as well as regulating cell-cell communication and signaling. Thus, EVs are important in intercellular communication and this is not limited to sister cells, but may also mediate the crosstalk between different cell types even over long distances. EVs play crucial functions in both cellular homeostasis and the pathogenesis of diseases, and since their contents reflect the status of the donor cell, they represent an additional valuable source of information for characterizing complex biological processes. Recent advances in isolation and analytical methods have led to substantial improvements in both characterizing and engineering EVs, leading to their use either as novel biomarkers for disease diagnosis/prognosis or even as novel therapies. Due to their capacity to carry biomolecules, various EV-based therapeutic applications have been devised for several pathological conditions, including eye diseases. In the eye, EVs have been detected in the retina, aqueous humor, vitreous body and also in tears. Experiences with other forms of intraocular drug applications have opened new ways to use EVs in the treatment of retinal diseases. We here provide a comprehensive summary of the main in vitro, in vivo, and ex vivo literature-based studies on EVs' role in ocular physiological and pathological conditions. We have focused on age-related macular degeneration, diabetic retinopathy, glaucoma, which are common eye diseases leading to permanent blindness, if not treated properly. In addition, the putative use of EVs in retinitis pigmentosa and other retinopathies is discussed. Finally, we have reviewed the potential of EVs as therapeutic tools and/or biomarkers in the above-mentioned retinal disorders. Evidence emerging from experimental disease models and human material strongly suggests future diagnostic and/or therapeutic exploitation of these biological agents in various ocular disorders with a good possibility to improve the patient's quality of life.
Collapse
Affiliation(s)
- Federico Manai
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Adrian Smedowski
- Department of Ophthalmology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland; GlaucoTech Co., Katowice, Poland
| | - Kai Kaarniranta
- Department of Ophthalmology, University of Eastern Finland, Kuopio, Finland; Department of Ophthalmology, Kuopio University Hospital, Kuopio, Finland; Department of Molecular Genetics, University of Lodz, Lodz, Poland
| | - Sergio Comincini
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | | |
Collapse
|
26
|
Liu Z, Zhang J, Li X, Hu Q, Chen X, Luo L, Ai L, Ye J. Astrocytic expression of Yes-associated protein (YAP) regulates retinal neovascularization in a mouse model of oxygen-induced retinopathy. Microvasc Res 2024; 151:104611. [PMID: 37774941 DOI: 10.1016/j.mvr.2023.104611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 09/19/2023] [Accepted: 09/19/2023] [Indexed: 10/01/2023]
Abstract
Pathological neovascularization is the hallmark of many vascular oculopathies. There is still a great deal of uncertainty surrounding retinal neovascularization research. A working hypothesis that astrocytic Yes-associated protein (YAP) act as a key factor in retinal neovascularization was proposed. And our study was conducted to verified this hypothesis. In vivo, we successfully generated mice deficient in YAP in astrocytes (YAPf/f GFAP-Cre mice) and set up oxygen-induced retinopathy (OIR) model. Pathological neovascularization was evaluated by immunofluorescence staining and western blotting. In vitro, cultured retinal astrocytes were transfected with YAP siRNA. Enzyme-linked immunosorbent assay (ELISA) and western blot were used to determine the proteins in the supernatants and cells. The results showed that YAP was upregulated and activated in the OIR mice retinas. Conditional ablation of YAP aggravated pathological neovascularization, along with the upregulation of vascular endothelial growth factor A (VEGF-A) and monocyte chemoattractant protein-1 (MCP-1). Studies in vitro confirmed that the knockdown of YAP in astrocytes lead to increases in VEGF-A and MCP-1 levels, thus enhancing pro-angiogenic capability of YAP-deficit astrocytes. In conclusion, astrocytic YAP alleviates retinal pathological angiogenesis by inhibiting the over-activation of astrocytes, which suppresses excessive VEGF-A production and neuroinflammation.
Collapse
Affiliation(s)
- Zhifei Liu
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Jieqiong Zhang
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Xue Li
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Qiumei Hu
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Xi Chen
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Linlin Luo
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Liqianyu Ai
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China.
| | - Jian Ye
- Department of Ophthalmology, Research Institute of Surgery & Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China.
| |
Collapse
|
27
|
Guan X, Shi C, Wang Y, He Y, Li Y, Yang Y, Mu W, Li W, Hou T. The possible role of Gremlin1 in inflammatory apical periodontitis. Arch Oral Biol 2024; 157:105848. [PMID: 37977053 DOI: 10.1016/j.archoralbio.2023.105848] [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: 09/21/2023] [Revised: 11/04/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023]
Abstract
OBJECTIVE In this study, we investigated the involvement of Gremlin1 on the pathological process of apical periodontitis and detect the underlying mechanisms preliminarily. METHODS Clinical healthy and inflamed periapical specimens were collected. Then, apical periodontitis (AP) animal models were established by consistent pulp exposure. In addition, AAV-shGremlin1 was injected into inflamed periapical lesions to inhibit the expression of Gremlin1. Alveolar bone loss was measured by Micro-CT. Furthermore, immunohistochemical or immunofluorescence staining of Gremlin1, phosphorylated-CREB, ICAM-1, VCAM-1, IL-1β were performed. RESULTS The expression of Gremlin1 is markedly increased in periapical lesions not only in clinic samples but also in animal models. Moreover, in rats' AP model, we uncovered that the Gremlin1 protein expression levels in apical lesions is positively correlated with those of IL-1β. Besides, the blockade of Gremlin1 in periapical lesions could substantially suppress the alveolar bone loss and restrains the inflammatory status by impacting the activation levels of phosphorylated-CREB, ICAM-1, VCAM-1, IL-1β. CONCLUSIONS Taken together, these results illustrated that Gremlin1 acts as a crucial mediator and possibly serves as a potential diagnostic marker for periapical periodontitis. Discovery of new factors involved in the pathophysiology of periapical periodontitis could contribute to the development of novel therapeutic treatment for the disease.
Collapse
Affiliation(s)
- Xiaoyue Guan
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xian Jiaotong University, Xi'an, Shaanxi, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Chen Shi
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xian Jiaotong University, Xi'an, Shaanxi, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Sichuan Hospital of Stomatology, Chengdu, Sichuan, China
| | - Yuting Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xian Jiaotong University, Xi'an, Shaanxi, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yani He
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xian Jiaotong University, Xi'an, Shaanxi, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yingxue Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xian Jiaotong University, Xi'an, Shaanxi, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yao Yang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xian Jiaotong University, Xi'an, Shaanxi, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Wenli Mu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xian Jiaotong University, Xi'an, Shaanxi, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Wenlan Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xian Jiaotong University, Xi'an, Shaanxi, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Tiezhou Hou
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China; Clinical Research Center of Shaanxi Province for Dental and Maxillofacial Diseases, College of Stomatology, Xian Jiaotong University, Xi'an, Shaanxi, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| |
Collapse
|
28
|
Pooresmaeil F, Andi S, Hasannejad-Asl B, Takamoli S, Bolhassani A. Engineered exosomes: a promising vehicle in cancer therapy. Ther Deliv 2023; 14:775-794. [PMID: 38116620 DOI: 10.4155/tde-2023-0131] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2023] Open
Abstract
During the past few decades, researchers have attempted to discover an effective treatment for cancer. Exosomes are natural nanovesicles released by various cells and play a role in communication between cells. While natural exosomes have high clinical potential, their inherent limitations have prompted researchers to design exosomes with improved therapeutic properties. To achieve this purpose, researchers have undertaken exosome engineering to modify the surface properties or internal composition of exosomes. After these modifications, engineered exosomes can be used as carriers for delivery of chemotherapeutic agents, targeted drug delivery or development of cancer vaccines. The present study provides an overview of exosomes, including their biogenesis, biological functions, isolation techniques, engineering methods, and potential applications in cancer therapy.
Collapse
Affiliation(s)
- Farkhondeh Pooresmaeil
- Department of Hepatitis & AIDS, Pasteur Institute of Iran, Tehran, 1316943551, Iran
- Department of Medical Biotechnology, School of Allied Medicine, Iran University of Medical Science, Tehran, Iran
| | - Sahar Andi
- Department of Molecular Medicine, School of Medicine, Qazvin University of Medical Science, Qazvin, Iran
| | - Behnam Hasannejad-Asl
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti, University of Medical Sciences, Tehran, Iran
| | - Shahla Takamoli
- Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran
| | - Azam Bolhassani
- Department of Hepatitis & AIDS, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| |
Collapse
|
29
|
Peng X, Zhang T, Wu Y, Wang X, Liu R, Jin X. mPEG-CS-modified flexible liposomes-reinforced thermosensitive sol-gel reversible hydrogels for ocular delivery of multiple drugs with enhanced synergism. Colloids Surf B Biointerfaces 2023; 231:113560. [PMID: 37812861 DOI: 10.1016/j.colsurfb.2023.113560] [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: 07/21/2023] [Revised: 09/02/2023] [Accepted: 09/17/2023] [Indexed: 10/11/2023]
Abstract
Non-invasive drug delivery offers a safe treatment while improving patient compliance. However, due to the particular physiological structure of the ocular, long-term retention and sustained drug release of the drug delivery system is crucial. Herein, this study aimed to design mPEG-CS-modified flexible liposomes-reinforced thermosensitive sol-gel reversible hydrogels (mPEG-CS-FL-TSG) for the delivery of astragaloside IV (AS-IV) and tetramethylpyrazine (TMP) to treat age-related macular degeneration. In vitro biological properties of mPEG-CS-FL and mPEG-CS-FL-TSG showed that they could be successfully taken up by ARPE-19 cells, and the uptake rate of mPEG-CS-FL-TSG was higher. Not only that, the release rate of mPEG-CS-FL-TSG was slower. More significantly, the results showed that the cytotoxicity of mPEG-CS-FL-TSG was lower than that of mPEG-CS-FL. In vivo result revealed that the drug delivery system could prominently enhance the ocular bioavailability of AS-IV and TMP, which is the enhanced synergism of well-permeable liposome and slow-releasing hydrogel. In summary, the mPEG-CS-FL-TSG can compensate for the short retention time and sudden release of liposome, as well as the low drug penetration of hydrogel, in order to show great promise in the non-invasive delivery of multiple drugs for the treatment of posterior ocular diseases.
Collapse
Affiliation(s)
- Xingru Peng
- State Key Laboratory of Component‑based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Tingting Zhang
- State Key Laboratory of Component‑based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yujie Wu
- State Key Laboratory of Component‑based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiaoyu Wang
- State Key Laboratory of Component‑based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Rui Liu
- State Key Laboratory of Component‑based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Xin Jin
- Department of Health Services, Logistics University of People's Armed Police Force, Tianjin, Tianjin 300162, China.
| |
Collapse
|
30
|
Pan T, Wu Y, Zhang X, Wang J, Wang X, Gu Q, Xu C, Fan Y, Li X, Xie P, Liu Q, Hu Z. Lens epithelial cell-derived exosome inhibits angiogenesis in ocular pathological neovascularization through its delivery of miR-146a-5p. FASEB J 2023; 37:e23192. [PMID: 37682530 DOI: 10.1096/fj.202301020rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/24/2023] [Accepted: 08/29/2023] [Indexed: 09/09/2023]
Abstract
Abnormal ocular neovascularization, a major pathology of eye diseases, leads to severe visual loss. The role of lens epithelial cell (LEC)-derived exosomes (Lec-exo) is largely unknown. Thus, we aimed to investigate whether Lec-exo can inhibit abnormal ocular neovascularization and explore the possible mechanisms. In our study, we proved the first evidence that exosomes derived from LECs attenuated angiogenesis in both oxygen-induced retinopathy and laser-induced choroidal neovascularization mice models. Further in vitro experiments proved that Lec-exo inhibited proliferation, migration, and tube formation capability of human umbilical vein endothelial cells in high glucose condition. Further high-throughput miRNAs sequencing analysis detected that miR-146a-5p was enriched in Lec-exo. Mechanistically, exosomal miR-146a-5p was delivered to endothelial cells and bound to the NRAS coding sequence, which subsequently inactivated AKT/ERK signaling pathway. We successfully elucidated the function of Lec-exo in inhibiting abnormal ocular neovascularization, which may offer a promising strategy for treatment of abnormal ocular neovascularization.
Collapse
Affiliation(s)
- Ting Pan
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Ophthalmology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Yan Wu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xu Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jingfan Wang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xingxing Wang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qinyuan Gu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Changlin Xu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuanyuan Fan
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xinsheng Li
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Ophthalmology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
| | - Ping Xie
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qinghuai Liu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zizhong Hu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
31
|
Roerig J, Schulz-Siegmund M. Standardization Approaches for Extracellular Vesicle Loading with Oligonucleotides and Biologics. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301763. [PMID: 37287374 DOI: 10.1002/smll.202301763] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/13/2023] [Indexed: 06/09/2023]
Abstract
Extracellular vesicles (EVs) are widely recognized for their potential as drug delivery systems. EVs are membranous nanoparticles shed from cells. Among their natural features are their ability to shield cargo molecules against degradation and enable their functional internalization into target cells. Especially biological or bio-inspired large molecules (LMs), like nucleic acids, proteins, peptides, and others, may profit from encapsulation in EVs for drug delivery purposes. In the last years, a variety of loading protocols are explored for different LMs. The lack of standardization in the EV drug delivery field has impeded their comparability so far. Currently, the first reporting frameworks and workflows for EV drug loading are proposed. The aim of this review is to summarize these evolving standardization approaches and set recently developed methods into context. This will allow for enhanced comparability of future work on EV drug loading with LMs.
Collapse
Affiliation(s)
- Josepha Roerig
- Pharmaceutical Technology, Institute of Pharmacy, Medical Faculty, Leipzig University, 04317, Leipzig, Germany
| | - Michaela Schulz-Siegmund
- Pharmaceutical Technology, Institute of Pharmacy, Medical Faculty, Leipzig University, 04317, Leipzig, Germany
| |
Collapse
|
32
|
Shafiq M, Rafique M, Cui Y, Pan L, Do CW, Ho EA. An insight on ophthalmic drug delivery systems: Focus on polymeric biomaterials-based carriers. J Control Release 2023; 362:446-467. [PMID: 37640109 DOI: 10.1016/j.jconrel.2023.08.041] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 08/31/2023]
Abstract
Presently, different types of eye diseases, such as glaucoma, myopia, infection, and dry eyes are treated with topical eye drops. However, due to ocular surface barriers, eye drops require multiple administrations, which may cause several risks, thereby necessitating additional strategies. Some of the key characteristics of an ideal ocular drug delivery system are as follows: (a) good penetration into cornea, (b) high drug retention in the ocular tissues, (c) targetability to the desired regions of the eye, and (d) good bioavailability. It is worthy to note that the corneal epithelial tight junctions hinder the permeation of therapeutics through the cornea. Therefore, it is necessary to design nanocarriers that can overcome these barriers and enhance drug penetration into the inner parts of the eye. Moreover, intelligent multifunctional nanocarriers can be designed to include cavities, which may help encapsulate sufficient amount of the drug. In addition, nanocarriers can be modified with the targeting moieties. Different types of nanocarriers have been developed for ocular drug delivery applications, including emulsions, liposomes, micelles, and nanoparticles. However, these formulations may be rapidly cleared from the eye. The therapeutic use of the nanoparticles (NPs) is also hindered by the non-specific adsorption of proteins on NPs, which may limit their interaction with the cellular moieties or other targeted biological factors. Functional drug delivery systems (DDS), which can offer targeted ocular drug delivery while avoiding the non-specific protein adsorption could exhibit great potential. This could be further realized by the on-demand DDS, which can respond to the stimuli in a spatio-temporal fashion. The cell-mediated DDS offer another valuable platform for ophthalmological drug delivery.
Collapse
Affiliation(s)
- Muhammad Shafiq
- Department of Chemical Engineering, Faculty of Engineering, Graduate School, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan; United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Muhammad Rafique
- Key Laboratory of Bioactive Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yingkun Cui
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong, China
| | - Li Pan
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong, China; First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China
| | - Chi-Wai Do
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong, China; Research Institute of Smart Ageing (RISA), The Hong Kong Polytechnic University, Hong Kong, China; Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Hong Kong
| | - Emmanuel A Ho
- School of Pharmacy, University of Waterloo, Waterloo, Canada; Waterloo Institute for Nanotechnology, Waterloo, Canada; Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Hong Kong.
| |
Collapse
|
33
|
Liu Q, Li D, Pan X, Liang Y. Targeted therapy using engineered extracellular vesicles: principles and strategies for membrane modification. J Nanobiotechnology 2023; 21:334. [PMID: 37717008 PMCID: PMC10505332 DOI: 10.1186/s12951-023-02081-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 08/26/2023] [Indexed: 09/18/2023] Open
Abstract
Extracellular vesicles (EVs) are 30-150 nm membrane-bound vesicles naturally secreted by cells and play important roles in intercellular communication by delivering regulatory molecules such as proteins, lipids, nucleic acids and metabolites to recipient cells. As natural nano-carriers, EVs possess desirable properties such as high biocompatibility, biological barrier permeability, low toxicity, and low immunogenicity, making them potential therapeutic delivery vehicles. EVs derived from specific cells have inherent targeting capacity towards specific cell types, which is yet not satisfactory enough for targeted therapy development and needs to be improved. Surface modifications endow EVs with targeting abilities, significantly improving their therapeutic efficiency. Herein, we first briefly introduce the biogenesis, composition, uptake and function of EVs, and review the cargo loading approaches for EVs. Then, we summarize the recent advances in surface engineering strategies of EVs, focusing on the applications of engineered EVs for targeted therapy. Altogether, EVs hold great promise for targeted delivery of various cargos, and targeted modifications show promising effects on multiple diseases.
Collapse
Affiliation(s)
- Qisong Liu
- National Clinical Research Center for Infectious Diseases, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, China
- Department of Orthopaedics, The Second Affiliated Hospital of Shenzhen University (People's Hospital of Shenzhen Baoan District), China, Shenzhen, 518000, China
| | - Defeng Li
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518020, China
| | - Xiaohua Pan
- Department of Orthopaedics, The Second Affiliated Hospital of Shenzhen University (People's Hospital of Shenzhen Baoan District), China, Shenzhen, 518000, China.
| | - Yujie Liang
- Department of Orthopaedics, The Second Affiliated Hospital of Shenzhen University (People's Hospital of Shenzhen Baoan District), China, Shenzhen, 518000, China.
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen Institute of Mental Health, Shenzhen Mental Health Center, Shenzhen Clinical Research Center for Mental Disorders, Shenzhen, 518020, Guangdong, China.
| |
Collapse
|
34
|
Li J, Zhang Y, Dong PY, Yang GM, Gurunathan S. A comprehensive review on the composition, biogenesis, purification, and multifunctional role of exosome as delivery vehicles for cancer therapy. Biomed Pharmacother 2023; 165:115087. [PMID: 37392659 DOI: 10.1016/j.biopha.2023.115087] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/24/2023] [Accepted: 06/26/2023] [Indexed: 07/03/2023] Open
Abstract
All forms of life produce nanosized extracellular vesicles called exosomes, which are enclosed in lipid bilayer membranes. Exosomes engage in cell-to-cell communication and participate in a variety of physiological and pathological processes. Exosomes function via their bioactive components, which are delivered to target cells in the form of proteins, nucleic acids, and lipids. Exosomes function as drug delivery vehicles due to their unique properties of innate stability, low immunogenicity, biocompatibility, biodistribution, accumulation in desired tissues, low toxicity in normal tissues, and the stimulation of anti-cancer immune responses, and penetration capacity into distance organs. Exosomes mediate cellular communications by delivering various bioactive molecules including oncogenes, oncomiRs, proteins, specific DNA, messenger RNA (mRNA), microRNA (miRNA), small interfering RNA (siRNA), and circular RNA (circRNA). These bioactive substances can be transferred to change the transcriptome of target cells and influence tumor-related signaling pathways. After considering all of the available literature, in this review we discuss the biogenesis, composition, production, and purification of exosomes. We briefly review exosome isolation and purification techniques. We explore great-length exosomes as a mechanism for delivering a variety of substances, including proteins, nucleic acids, small chemicals, and chemotherapeutic drugs. We also talk about the benefits and drawbacks of exosomes. This review concludes with a discussion future perspective and challenges. We hope that this review will provide us a better understanding of the current state of nanomedicine and exosome applications in biomedicine.
Collapse
Affiliation(s)
- Jian Li
- Fujian Key Laboratory of Traditional Chinese Veterinary Medicine and Animal Health, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Ye Zhang
- Advanced Medical Research Institute, Shandong University, Jinan, Shandong 250014, China
| | - Pei-Yu Dong
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, China
| | - Guo-Ming Yang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, China
| | - Sangiliyandi Gurunathan
- Department of Biotechnology, Rathinam College of Arts and Science, Pollachi Road, Eachanari, Coimbatore, Tamil Nadu 641021, India.
| |
Collapse
|
35
|
Yoshida YG, Yan S, Xu H, Yang J. Novel Metal Nanomaterials to Promote Angiogenesis in Tissue Regeneration. ENGINEERED REGENERATION 2023; 4:265-276. [PMID: 37234753 PMCID: PMC10207714 DOI: 10.1016/j.engreg.2023.03.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023] Open
Abstract
Angiogenesis-the formation of new blood vessels from existing blood vessels-has drawn significant attention in medical research. New techniques have been developed to control proangiogenic factors to obtain desired effects. Two important research areas are 1) understanding cellular mechanisms and signaling pathways involved in angiogenesis and 2) discovering new biomaterials and nanomaterials with proangiogenic effects. This paper reviews recent developments in controlling angiogenesis in the context of regenerative medicine and wound healing. We focus on novel proangiogenic materials that will advance the field of regenerative medicine. Specifically, we mainly focus on metal nanomaterials. We also discuss novel technologies developed to carry these proangiogenic inorganic molecules efficiently to target sites. We offer a comprehensive overview by combining existing knowledge regarding metal nanomaterials with novel developments that are still being refined to identify new nanomaterials.
Collapse
Affiliation(s)
- Yuki G. Yoshida
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA
| | - Su Yan
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Hui Xu
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Jian Yang
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
36
|
Ramsay E, Lajunen T, Bhattacharya M, Reinisalo M, Rilla K, Kidron H, Terasaki T, Urtti A. Selective drug delivery to the retinal cells: Biological barriers and avenues. J Control Release 2023; 361:1-19. [PMID: 37481214 DOI: 10.1016/j.jconrel.2023.07.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 06/09/2023] [Accepted: 07/16/2023] [Indexed: 07/24/2023]
Abstract
Retinal drug delivery is a challenging, but important task, because most retinal diseases are still without any proper therapy. Drug delivery to the retina is hampered by the anatomical and physiological barriers resulting in minimal bioavailability after topical ocular and systemic administrations. Intravitreal injections are current method-of-choice in retinal delivery, but these injections show short duration of action for small molecules and low target bioavailability for many protein, gene based drugs and nanomedicines. State-of-art delivery systems are based on prolonged retention, controlled drug release and physical features (e.g. size and charge). However, drug delivery to the retina is not cell-specific and these approaches do not facilitate intracellular delivery of modern biological drugs (e.g. intracellular proteins, RNA based medicines, gene editing). In this focused review we highlight biological factors and mechanisms that form the basis for the selective retinal drug delivery systems in the future. Therefore, we are presenting current knowledge related to retinal membrane transporters, receptors and targeting ligands in relation to nanomedicines, conjugates, extracellular vesicles, and melanin binding. These issues are discussed in the light of retinal structure and cell types as well as future prospects in the field. Unlike in some other fields of targeted drug delivery (e.g. cancer research), selective delivery technologies have been rarely studied, even though cell targeted delivery may be even more feasible after local administration into the eye.
Collapse
Affiliation(s)
- Eva Ramsay
- Drug Research Programme, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00014 University of Helsinki, Finland
| | - Tatu Lajunen
- Drug Research Programme, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00014 University of Helsinki, Finland; School of Pharmacy, University of Eastern Finland, Yliopistonranta 1 C, 70211 Kuopio, Finland
| | - Madhushree Bhattacharya
- Drug Research Programme, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00014 University of Helsinki, Finland
| | - Mika Reinisalo
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1 C, 70211 Kuopio, Finland
| | - Kirsi Rilla
- School of Medicine, University of Eastern Finland, Yliopistonranta 1 C, 70211 Kuopio, Finland
| | - Heidi Kidron
- Drug Research Programme, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00014 University of Helsinki, Finland
| | - Tetsuya Terasaki
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1 C, 70211 Kuopio, Finland
| | - Arto Urtti
- Drug Research Programme, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00014 University of Helsinki, Finland; School of Pharmacy, University of Eastern Finland, Yliopistonranta 1 C, 70211 Kuopio, Finland.
| |
Collapse
|
37
|
Danilushkina AA, Emene CC, Barlev NA, Gomzikova MO. Strategies for Engineering of Extracellular Vesicles. Int J Mol Sci 2023; 24:13247. [PMID: 37686050 PMCID: PMC10488046 DOI: 10.3390/ijms241713247] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/14/2023] [Accepted: 08/17/2023] [Indexed: 09/10/2023] Open
Abstract
Extracellular vesicles (EVs) are membrane vesicles released by cells into the extracellular space. EVs mediate cell-to-cell communication through local and systemic transportation of biomolecules such as DNA, RNA, transcription factors, cytokines, chemokines, enzymes, lipids, and organelles within the human body. EVs gained a particular interest from cancer biology scientists because of their role in the modulation of the tumor microenvironment through delivering bioactive molecules. In this respect, EVs represent an attractive therapeutic target and a means for drug delivery. The advantages of EVs include their biocompatibility, small size, and low immunogenicity. However, there are several limitations that restrict the widespread use of EVs in therapy, namely, their low specificity and payload capacity. Thus, in order to enhance the therapeutic efficacy and delivery specificity, the surface and composition of extracellular vesicles should be modified accordingly. In this review, we describe various approaches to engineering EVs, and further discuss their advantages and disadvantages to promote the application of EVs in clinical practice.
Collapse
Affiliation(s)
- Anna A. Danilushkina
- Laboratory of Intercellular Communications, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420021 Kazan, Russia
| | - Charles C. Emene
- Laboratory of Intercellular Communications, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420021 Kazan, Russia
| | - Nicolai A. Barlev
- Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
- Department of Biomedicine, Nazarbayev University School of Medicine, Astana 001000, Kazakhstan
| | - Marina O. Gomzikova
- Laboratory of Intercellular Communications, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420021 Kazan, Russia
- Laboratory of Molecular Immunology, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| |
Collapse
|
38
|
Wang Z, Tan W, Li B, Zou J, Li Y, Xiao Y, He Y, Yoshida S, Zhou Y. Exosomal non-coding RNAs in angiogenesis: Functions, mechanisms and potential clinical applications. Heliyon 2023; 9:e18626. [PMID: 37560684 PMCID: PMC10407155 DOI: 10.1016/j.heliyon.2023.e18626] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 07/14/2023] [Accepted: 07/21/2023] [Indexed: 08/11/2023] Open
Abstract
Exosomes are extracellular vesicles that can be produced by most cells. Exosomes act as important intermediaries in intercellular communication, and participate in a variety of biological activities between cells. Non-coding RNAs (ncRNAs) usually refer to RNAs that do not encode proteins. Although ncRNAs have no protein-coding capacity, they are able to regulate gene expression at multiple levels. Angiogenesis is the formation of new blood vessels from pre-existing vessels, which is an important physiological process. However, abnormal angiogenesis could induce many diseases such as atherosclerosis, diabetic retinopathy and cancer. Many studies have shown that ncRNAs can stably exist in exosomes and play a wide range of physiological and pathological roles including regulation of angiogenesis. In brief, some specific ncRNAs can be enriched in exosomes secreted by cells and absorbed by recipient cells through the exosome pathway, thus activating relevant signaling pathways in target cells and playing a role in regulating angiogenesis. In this review, we describe the physiological and pathological functions of exosomal ncRNAs in angiogenesis, summarize their role in angiogenesis-related diseases, and illustrate potential clinical applications like novel drug therapy strategies and diagnostic markers in exosome research as inspiration for future investigations.
Collapse
Affiliation(s)
- Zicong Wang
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Wei Tan
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Bingyan Li
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Jingling Zou
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Yun Li
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Yangyan Xiao
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Yan He
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Shigeo Yoshida
- Department of Ophthalmology, Kurume University School of Medicine, Fukuoka, 830-0011, Japan
| | - Yedi Zhou
- Department of Ophthalmology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Clinical Research Center of Ophthalmic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| |
Collapse
|
39
|
Tian Y, Zhang T, Li J, Tao Y. Advances in development of exosomes for ophthalmic therapeutics. Adv Drug Deliv Rev 2023; 199:114899. [PMID: 37236425 DOI: 10.1016/j.addr.2023.114899] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/18/2023] [Accepted: 05/21/2023] [Indexed: 05/28/2023]
Abstract
Exosomes contain multiple bioactive molecules and maintain the connection between cells. Recent advances in exosome-based therapeutics have witnessed unprecedented opportunities in treating ophthalmic diseases, including traumatic diseases, autoimmune diseases, chorioretinal diseases and others. Utilization of exosomes as delivery vectors to encapsulate both drugs and therapeutic genes could yield higher efficacy and avoid the unnecessary immune responses. However, exosome-based therapies also come with some potential ocular risks. In this review, we first present a general introduction to exosomes. Then we provide an overview of available applications and discuss their potential risks. Moreover, we review recently reported exosomes as delivery vectors for ophthalmic diseases. Finally, we put forward future perspectives to grapple with its translation and underlying issues.
Collapse
Affiliation(s)
- Ying Tian
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, PR China
| | - Tao Zhang
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, PR China
| | - Jing Li
- Beijing Key Laboratory of DNA Damage Response, College of Life Sciences, Capital Normal University, Beijing 100048, PR China
| | - Yong Tao
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, PR China.
| |
Collapse
|
40
|
Li S, Chen L, Fu Y. Nanotechnology-based ocular drug delivery systems: recent advances and future prospects. J Nanobiotechnology 2023; 21:232. [PMID: 37480102 PMCID: PMC10362606 DOI: 10.1186/s12951-023-01992-2] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 07/09/2023] [Indexed: 07/23/2023] Open
Abstract
Ocular drug delivery has constantly challenged ophthalmologists and drug delivery scientists due to various anatomical and physiological barriers. Static and dynamic ocular barriers prevent the entry of exogenous substances and impede therapeutic agents' active absorption. This review elaborates on the anatomy of the eye and the associated constraints. Followed by an illustration of some common ocular diseases, including glaucoma and their current clinical therapies, emphasizing the significance of drug therapy in treating ocular diseases. Subsequently, advances in ocular drug delivery modalities, especially nanotechnology-based ocular drug delivery systems, are recommended, and some typical research is highlighted. Based on the related research, systematic and comprehensive characterizations of the nanocarriers are summarized, hoping to assist with future research. Besides, we summarize the nanotechnology-based ophthalmic drugs currently on the market or still in clinical trials and the recent patents of nanocarriers. Finally, inspired by current trends and therapeutic concepts, we provide an insight into the challenges faced by novel ocular drug delivery systems and further put forward directions for future research. We hope this review can provide inspiration and motivation for better design and development of novel ophthalmic formulations.
Collapse
Affiliation(s)
- Shiding Li
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, China
| | - Liangbo Chen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, China
| | - Yao Fu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, China.
| |
Collapse
|
41
|
Wu T, Jiang Y, Shi W, Wang Y, Li T. Endoplasmic reticulum stress: a novel targeted approach to repair bone defects by regulating osteogenesis and angiogenesis. J Transl Med 2023; 21:480. [PMID: 37464413 PMCID: PMC10353205 DOI: 10.1186/s12967-023-04328-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 07/06/2023] [Indexed: 07/20/2023] Open
Abstract
Bone regeneration therapy is clinically important, and targeted regulation of endoplasmic reticulum (ER) stress is important in regenerative medicine. The processing of proteins in the ER controls cell fate. The accumulation of misfolded and unfolded proteins occurs in pathological states, triggering ER stress. ER stress restores homeostasis through three main mechanisms, including protein kinase-R-like ER kinase (PERK), inositol-requiring enzyme 1ɑ (IRE1ɑ) and activating transcription factor 6 (ATF6), collectively known as the unfolded protein response (UPR). However, the UPR has both adaptive and apoptotic effects. Modulation of ER stress has therapeutic potential for numerous diseases. Repair of bone defects involves both angiogenesis and bone regeneration. Here, we review the effects of ER stress on osteogenesis and angiogenesis, with emphasis on ER stress under high glucose (HG) and inflammatory conditions, and the use of ER stress inducers or inhibitors to regulate osteogenesis and angiogenesis. In addition, we highlight the ability for exosomes to regulate ER stress. Recent advances in the regulation of ER stress mediated osteogenesis and angiogenesis suggest novel therapeutic options for bone defects.
Collapse
Affiliation(s)
- Tingyu Wu
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, 266003, China
| | - Yaping Jiang
- Department of Oral Implantology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Weipeng Shi
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, 266003, China
| | - Yingzhen Wang
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, 266003, China
| | - Tao Li
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, 266003, China.
| |
Collapse
|
42
|
Wang Y, Qin WY, Wang Q, Liu XN, Li XH, Ye XQ, Bai Y, Zhang Y, Liu P, Wang XL, Zhou YH, Shao ZB, Yuan HP. Young Sca-1 + bone marrow stem cell-derived exosomes preserve visual function via the miR-150-5p/MEKK3/JNK/c-Jun pathway to reduce M1 microglial polarization. J Nanobiotechnology 2023; 21:194. [PMID: 37322478 DOI: 10.1186/s12951-023-01944-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 05/29/2023] [Indexed: 06/17/2023] Open
Abstract
BACKGROUND Polarization of microglia, the resident retinal immune cells, plays important roles in mediating both injury and repair responses post-retinal ischemia-reperfusion (I/R) injury, which is one of the main pathological mechanisms behind ganglion cell apoptosis. Aging could perturb microglial balances, resulting in lowered post-I/R retinal repair. Young bone marrow (BM) stem cell antigen 1-positive (Sca-1+) cells have been demonstrated to have higher reparative capabilities post-I/R retinal injury when transplanted into old mice, where they were able to home and differentiate into retinal microglia. METHODS Exosomes were enriched from young Sca-1+ or Sca-1- cells, and injected into the vitreous humor of old mice post-retinal I/R. Bioinformatics analyses, including miRNA sequencing, was used to analyze exosome contents, which was confirmed by RT-qPCR. Western blot was then performed to examine expression levels of inflammatory factors and underlying signaling pathway proteins, while immunofluorescence staining was used to examine the extent of pro-inflammatory M1 microglial polarization. Fluoro-Gold labelling was then utilized to identify viable ganglion cells, while H&E staining was used to examine retinal morphology post-I/R and exosome treatment. RESULTS Sca-1+ exosome-injected mice yielded better visual functional preservation and lowered inflammatory factors, compared to Sca-1-, at days 1, 3, and 7 days post-I/R. miRNA sequencing found that Sca-1+ exosomes had higher miR-150-5p levels, compared to Sca-1- exosomes, which was confirmed by RT-qPCR. Mechanistic analysis found that miR-150-5p from Sca-1+ exosomes repressed the mitogen-activated protein kinase kinase kinase 3 (MEKK3)/JNK/c-Jun axis, leading to IL-6 and TNF-α downregulation, and subsequently reduced microglial polarization, all of which contributes to reduced ganglion cell apoptosis and preservation of proper retinal morphology. CONCLUSION This study elucidates a potential new therapeutic approach for neuroprotection against I/R injury, via delivering miR-150-5p-enriched Sca-1+ exosomes, which targets the miR-150-5p/MEKK3/JNK/c-Jun axis, thereby serving as a cell-free remedy for treating retinal I/R injury and preserving visual functioning.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wan-Yun Qin
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qi Wang
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry Education, Harbin, China
| | - Xin-Na Liu
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry Education, Harbin, China
| | - Xiang-Hui Li
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xin-Qi Ye
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ying Bai
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yan Zhang
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Pan Liu
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xin-Lin Wang
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yu-Hang Zhou
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zheng-Bo Shao
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Hui-Ping Yuan
- Department of Ophthalmology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
43
|
Yao X, Xue Y, Ma Q, Bai Y, Jia P, Zhang Y, Lai B, He S, Ma Q, Zhang J, Tian H, Yin Q, Zheng X, Zheng X. 221S-1a inhibits endothelial proliferation in pathological angiogenesis through ERK/c-Myc signaling. Eur J Pharmacol 2023:175805. [PMID: 37247812 DOI: 10.1016/j.ejphar.2023.175805] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 05/12/2023] [Accepted: 05/22/2023] [Indexed: 05/31/2023]
Abstract
Pathological angiogenesis plays a major role in many disease processes, including cancer and diabetic retinopathy. Antiangiogenic therapy is a potential management for pathologic angiogenesis. The novel synthetic compound 221S-1a, derived from captopril, tanshinol and borneol, may have antiangiogenic properties. On the basis of MS, NMR and HPLC analysis, the structure of 221S-1a was identified. The cellular uptake and metabolism of this compound was also observed. Next, the antiangiogenic properties of 221S-1a were evaluated in tumor-xenograft and OIR models in vivo. The inhibitory properties of 221S-1a on endothelial cell proliferation, migration, tube formation and sprouting were detected in vitro. Furthermore, 221S-1a induced G1/S phase arrest was detected by PI staining flow cytometry analysis and Cyclin D, Cyclin E expression. 221S-1a inhibited ERK1/2 activation and nuclear translocation, in addition to downregulation of c-Myc, a transcription factor that regulates cell cycle progression. Molecular docking indicated the interaction of 221S-1a with the ATP-binding site of ERK2, leading to the inhibition of ERK2 phosphorylation and a concomitant inhibition of ERK1 phosphorylation. In conclusion, 221S-1a inhibited the G1/S phase transition by blocking the ERK1/2/c-Myc pathway to reduce tumor and OIR retinal angiogenesis. These novel findings suggest that 221S-1a is a potential pharmacologic candidate for treating pathological angiogenesis.
Collapse
Affiliation(s)
- Xinye Yao
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Yanbo Xue
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qiang Ma
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Department of Peripheral Vascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yajun Bai
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, China
| | - Pu Jia
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, China
| | - Yiman Zhang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Department of Peripheral Vascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Baochang Lai
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China; Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education of China, Xi'an, China
| | - Shuting He
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Qiong Ma
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Junbo Zhang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Department of Peripheral Vascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hongyan Tian
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Department of Peripheral Vascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qian Yin
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Department of Peripheral Vascular, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China.
| | - Xiaohui Zheng
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, China.
| | - Xiaopu Zheng
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
44
|
Icariside II alleviates ischemic retinopathy by modulating microglia and promoting vessel integrity. J Funct Foods 2023. [DOI: 10.1016/j.jff.2023.105510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2023] Open
|
45
|
Zhao L, Ling L, Lu J, Jiang F, Sun J, Zhang Z, Huang Y, Liu X, Zhu Y, Fu X, Peng S, Yuan W, Zhao R, Zhang Z. Reactive oxygen species-responsive mitochondria-targeted liposomal quercetin attenuates retinal ischemia-reperfusion injury via regulating SIRT1/FOXO3A and p38 MAPK signaling pathways. Bioeng Transl Med 2023; 8:e10460. [PMID: 37206232 PMCID: PMC10189480 DOI: 10.1002/btm2.10460] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 11/15/2022] [Accepted: 11/17/2022] [Indexed: 12/03/2022] Open
Abstract
Retinal ischemia-reperfusion (RIR) injury is involved in the pathogenesis of various vision-threatening diseases. The overproduction of reactive oxygen species (ROS) is thought to be the main cause of RIR injury. A variety of natural products, including quercetin (Que), exhibit potent antioxidant activity. However, the lack of an efficient delivery system for hydrophobic Que and the presence of various intraocular barriers limit the effective retinal delivery of Que in clinical settings. In this study, we encapsulated Que into ROS-responsive mitochondria-targeted liposomes (abbreviated to Que@TPP-ROS-Lips) to achieve the sustained delivery of Que to the retina. The intracellular uptake, lysosome escape ability, and mitochondria targeting ability of Que@TPP-ROS-Lips were evaluated in R28 retinal cells. Treating R28 cells with Que@TPP-ROS-Lips significantly ameliorated the decrease in ATP content, ROS generation, and increase in the release of lactate dehydrogenase in an in vitro oxygen-glucose deprivation (OGD) model of retinal ischemia. In a rat model, the intravitreal injection of Que@TPP-ROS-Lips 24 h after inducing retinal ischemia significantly enhanced retinal electrophysiological recovery and reduced neuroinflammation, oxidative stress, and apoptosis. Que@TPP-ROS-Lips were taken up by retina for at least 14 days after intravitreal administration. Molecular docking and functional biological experiments revealed that Que targets FOXO3A to inhibit oxidative stress and inflammation. Que@TPP-ROS-Lips also partially inhibited the p38 MAPK signaling pathway, which contributes to oxidative stress and inflammation. In conclusion, our new platform for ROS-responsive and mitochondria-targeted drug release shows promise for the treatment of RIR injury and promotes the clinical application of hydrophobic natural products.
Collapse
Affiliation(s)
- Laien Zhao
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of ShandongYantai UniversityYantaiPeople's Republic of China
| | - Longbing Ling
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of ShandongYantai UniversityYantaiPeople's Republic of China
| | - Jing Lu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of ShandongYantai UniversityYantaiPeople's Republic of China
| | - Feng Jiang
- Department of OphthalmologyTianjin Medical University General HospitalTianjinPeople's Republic of China
| | - Jianchao Sun
- School of Environment and Material EngineeringYantai UniversityYantaiPeople's Republic of China
| | - Zhen Zhang
- College of Chemistry and Chemical EngineeringYantai UniversityYantaiPeople's Republic of China
| | - Yanmei Huang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of ShandongYantai UniversityYantaiPeople's Republic of China
| | - Xiaoqian Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of ShandongYantai UniversityYantaiPeople's Republic of China
| | - Yanjuan Zhu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of ShandongYantai UniversityYantaiPeople's Republic of China
| | - Xiaoxuan Fu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of ShandongYantai UniversityYantaiPeople's Republic of China
| | - Shengjun Peng
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of ShandongYantai UniversityYantaiPeople's Republic of China
| | - Wenze Yuan
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of ShandongYantai UniversityYantaiPeople's Republic of China
| | - Ruikang Zhao
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of ShandongYantai UniversityYantaiPeople's Republic of China
| | - Zhuhong Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of ShandongYantai UniversityYantaiPeople's Republic of China
| |
Collapse
|
46
|
Chatterjee A, Singh R. Extracellular vesicles: an emerging player in retinal homeostasis. Front Cell Dev Biol 2023; 11:1059141. [PMID: 37181750 PMCID: PMC10166895 DOI: 10.3389/fcell.2023.1059141] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 04/10/2023] [Indexed: 05/16/2023] Open
Abstract
Extracellular vesicles (EVs) encompass secreted membrane vesicles of varied sizes, including exosomes (-30-200 nm) and microvesicles (MVs) that are ∼100-1,000 nm in size. EVs play an important role in autocrine, paracrine, and endocrine signaling and are implicated in myriad human disorders including prominent retinal degenerative diseases, like age related macular degeneration (AMD) and diabetic retinopathy (DR). Studies of EVs in vitro using transformed cell lines, primary cultures, and more recently, induced pluripotent stem cell derived retinal cell type(s) (e.g., retinal pigment epithelium) have provided insights into the composition and function of EVs in the retina. Furthermore, consistent with a causal role of EVs in retinal degenerative diseases, altering EV composition has promoted pro-retinopathy cellular and molecular events in both in vitro and in vivo models. In this review, we summarize the current understanding of the role of EVs in retinal (patho)physiology. Specifically, we will focus on disease-associated EV alterations in specific retinal diseases. Furthermore, we discuss the potential utility of EVs in diagnostic and therapeutic strategies for targeting retinal diseases.
Collapse
Affiliation(s)
- Amit Chatterjee
- Department of Ophthalmology, University of Rochester, Rochester, NY, United States
- Department of Biomedical Genetics, University of Rochester, Rochester, NY, United States
- Center for Visual Science, University of Rochester, Rochester, NY, United States
| | - Ruchira Singh
- Department of Ophthalmology, University of Rochester, Rochester, NY, United States
- Department of Biomedical Genetics, University of Rochester, Rochester, NY, United States
- Center for Visual Science, University of Rochester, Rochester, NY, United States
- UR Stem Cell and Regenerative Medicine Center, University of Rochester, Rochester, NY, United States
| |
Collapse
|
47
|
Shekari F, Abyadeh M, Meyfour A, Mirzaei M, Chitranshi N, Gupta V, Graham SL, Salekdeh GH. Extracellular Vesicles as reconfigurable therapeutics for eye diseases: Promises and hurdles. Prog Neurobiol 2023; 225:102437. [PMID: 36931589 DOI: 10.1016/j.pneurobio.2023.102437] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 03/14/2023] [Indexed: 03/17/2023]
Abstract
A large number of people worldwide suffer from visual impairment. However, most available therapies rely on impeding the development of a particular eye disorder. Therefore, there is an increasing demand for effective alternative treatments, specifically regenerative therapies. Extracellular vesicles, including exosomes, ectosomes, or microvesicles, are released by cells and play a potential role in regeneration. Following an introduction to EV biogenesis and isolation methods, this integrative review provides an overview of our current knowledge about EVs as a communication paradigm in the eye. Then, we focused on the therapeutic applications of EVs derived from conditioned medium, biological fluid, or tissue and highlighted some recent developments in strategies to boost the innate therapeutic potential of EVs by loading various kinds of drugs or being engineered at the level of producing cells or EVs. Challenges faced in the development of safe and effective translation of EV-based therapy into clinical settings for eye diseases are also discussed to pave the road toward reaching feasible regenerative therapies required for eye-related complications.
Collapse
Affiliation(s)
- Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Advanced Therapy Medicinal Product Technology Development Center (ATMP-TDC), Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | | | - Anna Meyfour
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Mirzaei
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW, Australia
| | - Nitin Chitranshi
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW, Australia
| | - Vivek Gupta
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW, Australia
| | - Stuart L Graham
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, North Ryde, Sydney, NSW, Australia
| | | |
Collapse
|
48
|
Xiao G, Xu Z, Luo F. Combinational antitumor strategies of exosomes as drug carriers: Mini review. Front Pharmacol 2023; 13:1107329. [PMID: 36744207 PMCID: PMC9897293 DOI: 10.3389/fphar.2022.1107329] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 12/28/2022] [Indexed: 01/21/2023] Open
Abstract
Cancer therapies have made tremendous progress in the last decade, but monotherapy still has apparent limitations and lacks therapeutic efficacy. Thus, the simultaneous administration of multiple drugs has been widely explored and has shown better outcomes. Exosomes, deriving from almost all living cells, are natural nanocarriers designed to deliver drugs to tumor sites. Therefore, combinational antitumor therapies based on exosomes, such as engineered exosomes and different combinations of chemotherapeutic agents, therapeutic nucleic acids, photosensitizers, immunotherapy and phytochemicals, have considerable prospects and potential for clinical translation. Here, we summarize current strategies of cancer combination therapy in exosomes and propose opportunities and challenges in the future.
Collapse
Affiliation(s)
- Guixiu Xiao
- Lung Cancer Center, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Zihan Xu
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Feng Luo
- Lung Cancer Center, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, Sichuan, China,*Correspondence: Feng Luo,
| |
Collapse
|
49
|
Small Extracellular Vesicles as a New Class of Medicines. Pharmaceutics 2023; 15:pharmaceutics15020325. [PMID: 36839647 PMCID: PMC9961868 DOI: 10.3390/pharmaceutics15020325] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
Extracellular vesicles (EVs) are nanovesicles that are naturally released from cells in a lipid bilayer-bound form. A subset population with a size of 200 nm, small EVs (sEVs), is enticing in many ways. Initially perceived as mere waste receptacles, sEVs have revealed other biological functions, such as cell-to-cell signal transduction and communication. Besides their notable biological functions, sEVs have profound advantages as future drug modalities: (i) excellent biocompatibility, (ii) high stability, and (iii) the potential to carry undruggable macromolecules as cargo. Indeed, many biopharmaceutical companies are utilizing sEVs, not only as diagnostic biomarkers but as therapeutic drugs. However, as all inchoate fields are challenging, there are limitations and hindrances in the clinical translation of sEV therapeutics. In this review, we summarize different types of sEV therapeutics, future improvements, and current strategies in large-scale production.
Collapse
|
50
|
Calpain-2 Facilitates Autophagic/Lysosomal Defects and Apoptosis in ARPE-19 Cells and Rats Induced by Exosomes from RPE Cells under NaIO 3 Stimulation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:3310621. [PMID: 36703913 PMCID: PMC9873447 DOI: 10.1155/2023/3310621] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 10/07/2022] [Accepted: 12/31/2022] [Indexed: 01/19/2023]
Abstract
Although accumulated evidence supports the notion that calpain contributes to eye disease, the mechanisms by which calpain promotes RPE injury are not defined. The present study is aimed at investigating whether the effect of NaIO3-exos (exosomes derived from RPE cells under NaIO3 stimulation) on the dysfunction of the autophagy-lysosomal pathway (ALP) and apoptosis is based on its regulation of calpain activation in ARPE-19 cells and rats. The results showed that calpain-2 activation, ALP dysfunction, and apoptosis were induced by NaIO3-exos in ARPE-19 cells. NaIO3-exo significantly increased autophagic substrates by activating lysosomal dysfunction. ALP dysfunction and apoptosis in vitro could be eliminated by knocking down calpain-2 (si-C2) or the inhibitor calpain-2-IN-1. Further studies indicated that NaIO3-exo enhanced calpain-2 expression, ALP dysfunction, apoptosis, and retinal damage in rats. In summary, these results demonstrate for the first time that calpain-2 is one of the key players in the NaIO3-exo-mediated ALP dysfunction, apoptosis, and retinal damage and identify calpain-2 as a promising target for therapies aimed at age-related macular degeneration (AMD).
Collapse
|