1
|
Chen Q, Zheng Y, Jiang X, Wang Y, Chen Z, Wu D. Nature's carriers: leveraging extracellular vesicles for targeted drug delivery. Drug Deliv 2024; 31:2361165. [PMID: 38832506 DOI: 10.1080/10717544.2024.2361165] [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: 08/07/2023] [Accepted: 05/14/2024] [Indexed: 06/05/2024] Open
Abstract
With the rapid development of drug delivery systems, extracellular vesicles (EVs) have emerged as promising stars for improving targeting abilities and realizing effective delivery. Numerous studies have shown when compared to conventional strategies in targeted drug delivery (TDD), EVs-based strategies have several distinguished advantages besides targeting, such as participating in cell-to-cell communications and immune response, showing high biocompatibility and stability, penetrating through biological barriers, etc. In this review, we mainly focus on the mass production of EVs including the challenges and strategies for scaling up EVs production in a cost-effective and reproducible manner, the loading and active targeting methods, and examples of EVs as vehicles for TDD in consideration of potential safety and regulatory issues associated. We also conclude and discuss the rigor and reproducibility of EVs production, the current research status of the application of EVs-based strategies to targeted drug delivery, clinical conversion prospects, and existing chances and challenges.
Collapse
Affiliation(s)
- Qi Chen
- Interdisciplinary Institute for Medical Engineering, Fuzhou University, Fuzhou, P. R. China
| | - Yuyi Zheng
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xuhong Jiang
- Epilepsy Center, Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Yi Wang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Rehabilitation Medical Center, The Third Affiliated Hospital of Zhejiang, Chinese Medical University, Hangzhou, PR China
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Epilepsy Center, Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Di Wu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
2
|
Jiang Q, Wang L, Tian J, Zhang W, Cui H, Gui H, Zang Z, Li B, Si X. Food-derived extracellular vesicles: natural nanocarriers for active phytoconstituents in new functional food. Crit Rev Food Sci Nutr 2024; 64:11701-11721. [PMID: 37548408 DOI: 10.1080/10408398.2023.2242947] [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: 08/08/2023]
Abstract
Extracellular vesicles (EVs) are naturally occurring non-replicating particles released from cells, known for their health-promoting effects and potential as carriers for drug delivery. Extensive research has been conducted on delivery systems based on culture-cell-derived EVs. Nevertheless, they have several limitations including low production yield, high expenses, unsuitability for oral administration, and safety concerns in applications. Conversely, food-derived EVs (FDEVs) offer unique advantages that cannot be easily substituted. This review provides a comprehensive analysis of the biogenesis pathways, composition, and health benefits of FDEVs, as well as the techniques required for constructing oral delivery systems. Furthermore, it explores the advantages and challenges associated with FDEVs as oral nanocarriers, and discusses the current research advancements in delivering active phytoconstituents. FDEVs, functioning as a nanocarrier platform for the oral delivery of active molecules, present numerous benefits such as convenient administration, high biocompatibility, low toxicity, and inherent targeting. Nevertheless, numerous unresolved issues persist in the isolation, characterization, drug loading, and application of FDEVs. Technical innovation and standardization of quality control are the key points to promote the development of FDEVs. The review aimed to provide frontier ideas and basic quality control guidelines for developing new functional food based on FDEVs oral drug delivery system.
Collapse
Affiliation(s)
- Qiao Jiang
- College of Food Science, Shenyang Agricultural University, Shenyang, China
| | - Li Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Jinlong Tian
- College of Food Science, Shenyang Agricultural University, Shenyang, China
| | - Weijia Zhang
- College of Food Science, Shenyang Agricultural University, Shenyang, China
| | - Huijun Cui
- College of Food Science, Shenyang Agricultural University, Shenyang, China
| | - Hailong Gui
- College of Food Science, Shenyang Agricultural University, Shenyang, China
| | - Zhihuan Zang
- College of Food Science, Shenyang Agricultural University, Shenyang, China
| | - Bin Li
- College of Food Science, Shenyang Agricultural University, Shenyang, China
| | - Xu Si
- College of Food Science, Shenyang Agricultural University, Shenyang, China
| |
Collapse
|
3
|
Palakurthi SS, Shah B, Kapre S, Charbe N, Immanuel S, Pasham S, Thalla M, Jain A, Palakurthi S. A comprehensive review of challenges and advances in exosome-based drug delivery systems. NANOSCALE ADVANCES 2024:d4na00501e. [PMID: 39484149 PMCID: PMC11523810 DOI: 10.1039/d4na00501e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 09/22/2024] [Indexed: 11/03/2024]
Abstract
Exosomes or so-called natural nanoparticles have recently shown enormous potential for targeted drug delivery systems. Several studies have reported that exosomes as advanced drug delivery platforms offer efficient targeting of chemotherapeutics compared to individual polymeric nanoparticles or liposomes. Taking structural constituents of exosomes, viz., proteins, nucleic acids, and lipids, into consideration, exosomes are the most promising carriers as genetic messengers and for treating genetic deficiencies or tumor progression. Unfortunately, very little attention has been paid to the factors like source, scalability, stability, and validation that contribute to the quality attributes of exosome-based drug products. Some studies suggested that exosomes were stable at around -80 °C, which is impractical for storing pharmaceutical products. Currently, no reports on the shelf-life and in vivo stability of exosome formulations are available. Exosomes are quickly cleared from blood circulation, and their in vivo distribution depends on the source. Considering these challenges, further studies are necessary to address major limitations such as poor drug loading, reduced in vivo stability, a need for robust, economical, and scalable production methods, etc., which may unlock the potential of exosomes in clinical applications. A few reports based on hybrid exosomes involving hybridization between different cell/tumor/macrophage-derived exosomes with synthetic liposomes through membrane fusion have shown to overcome some limitations associated with natural or synthetic exosomes. Yet, sufficient evidence is indispensable to prove their stability and clinical efficacy.
Collapse
Affiliation(s)
- Sushesh Srivatsa Palakurthi
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University Kingsville TX 78363 USA +1-361-221-0748
| | - Brijesh Shah
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University Kingsville TX 78363 USA +1-361-221-0748
| | - Sumedha Kapre
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University Kingsville TX 78363 USA +1-361-221-0748
| | - Nitin Charbe
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University Kingsville TX 78363 USA +1-361-221-0748
| | - Susan Immanuel
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University Kingsville TX 78363 USA +1-361-221-0748
| | - Sindhura Pasham
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University Kingsville TX 78363 USA +1-361-221-0748
| | - Maharshi Thalla
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University Kingsville TX 78363 USA +1-361-221-0748
| | - Ankit Jain
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University Kingsville TX 78363 USA +1-361-221-0748
| | - Srinath Palakurthi
- Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University Kingsville TX 78363 USA +1-361-221-0748
| |
Collapse
|
4
|
Wang M, Yang X, Ye Y, Fan K, Chen C, Zheng L, Li X, Dong C, Li C, Dong N. Anti-inflammatory and Restorative effects of milk exosomes and Dexamethasone-Loaded exosomes in a corneal alkali burn model. Int J Pharm 2024; 666:124784. [PMID: 39357813 DOI: 10.1016/j.ijpharm.2024.124784] [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: 01/21/2024] [Revised: 09/11/2024] [Accepted: 09/29/2024] [Indexed: 10/04/2024]
Abstract
Corneal alkali burn is a common and challenging ocular trauma, necessitating the use of dexamethasone (DXMS) as a therapeutic agent. However, prolonged and frequent administration of this drug can lead to undesirable side effects, limiting its clinical application. This study aimed to investigate the role and mechanism of action of exosomes as drug carriers in corneal alkali burn repair. We employed centrifugation to isolate milk exosomes (EXO) as nanocarriers. We observed that EXO enhanced the activity and migration of corneal epithelial cells, expediting the repair process following corneal injury. Additionally, a nano-drug delivery model (DXMS@EXO) was designed using ultrasound to load DXMS into exosomes, thus enabling targeted delivery to inflammatory cells and enhancing drug efficacy. DXMS@EXO inhibited the inflammatory processes in the corneal alkali burn model by modulating the classical Wnt signaling pathway, thereby promoting corneal re-epithelialization and wound healing and accelerating the repair process of corneal alkali burn. Neither EXO nor DXMS@EXO exhibited significant side effects during the course of treatment. This study highlighted the substantial potential of EXO and DXMS@EXO in improving drug efficacy and facilitating the repair of corneal alkali burn.
Collapse
Affiliation(s)
- Mengyuan Wang
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine & School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Fujian, China
| | - Xiuqin Yang
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine & School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Fujian, China
| | - Yingyue Ye
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine & School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Fujian, China
| | - Kai Fan
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine & School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Fujian, China
| | - Cuiting Chen
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine & School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Fujian, China
| | - Lan Zheng
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine & School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Fujian, China
| | - Xiang Li
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine & School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Fujian, China
| | - Chenyu Dong
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine & School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Cheng Li
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine & School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, School of Medicine, Xiamen University, Fujian, China.
| | - Nuo Dong
- Eye Institute & Affiliated Xiamen Eye Center, School of Medicine & School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China; Xiamen Clinical Research Center for Eye Diseases, Xiamen, Fujian, China; Xiamen Key Laboratory of Ophthalmology, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Ocular Surface and Corneal Disease, Xiamen, Fujian, China; Xiamen Key Laboratory of Corneal & Ocular Surface Diseases, Xiamen, Fujian, China; Translational Medicine Institute of Xiamen Eye Center of Xiamen University, Xiamen, Fujian, China; Huaxia Eye Hospital of Quanzhou, Quanzhou, Fujian, China; Department of Ophthalmology, Affiliated People's Hospital & Zhenjiang Kangfu Eye Hospital, Zhenjiang College, Zhenjiang, Jiangsu, China.
| |
Collapse
|
5
|
Aare M, Bagde A, Nathani A, Rishi AK, Singh M. Enhanced oral bioavailability and in vitro evaluation of cannabidiol camel milk-derived exosome formulation in resistant MDA-MB-231 and MDA-MB-468 breast cancer cells. Int J Pharm 2024; 663:124375. [PMID: 38914353 DOI: 10.1016/j.ijpharm.2024.124375] [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: 03/12/2024] [Revised: 06/17/2024] [Accepted: 06/20/2024] [Indexed: 06/26/2024]
Abstract
The potential of camel milk-derived exosomes (CMDE) to enhance the bioavailability of Cannabidiol (CBD) was investigated. CBD-CMDE formulation was prepared using an established procedure and its particle size was 138.4 ± 4.37 nm, and CBD entrapment efficiency of 56.56 ± 4.26 %. In-vitro release studies showed release of 78.27 ± 5.37 % and 46.42 ± 4.75 % CBD from CMDE and control CBD formulation respectively in pH 6.8 at 24 hr. The apparent permeability (Papp) of CBD-CMDE was found to be enhanced by 3.95-fold with Papp of 22.9*10-6 ± 0.34 cm/sec as compared to control CBD formulation with Papp of 5.8*10-6 ± 0.65 cm/sec in MDCK cells. CBD-CMDE was found to be more potent than CBD in 2D cytotoxicity assay with IC50 values of 3.6 ± 0.54 µM, 3.88 ± 0.54 µM and 7.53 ± 0.59 µM, 7.53 ± 0.59 µM against Doxorubicin (DOX) resistant MDA-MB-231 and Rapamycin (RM) resistant MDA-MB-468 breast cancer cells respectively. Moreover, 3D spheroids assay results demonstrated CBD-CMDE with IC50 values of 14 ± 0.85 µM, 15 ± 0.07 µM as compared to CBD alone with IC50 values of 25 ± 0.93 µM, 34.7 ± 0.08 µM in MDA-MB-231 DOX RT cells and MDA-MB-468 RM RT cells respectively. In-vivo PK studies showed enhanced bioavailability of CBD from CBD-exosomes with AUC(0-24h) of 1350.56 ± 187.50 h.ng/mL as compared to CBD control formulation with AUC(0-24h) of 351.95 ± 39.10 h.ng/mL with a single oral dose of 12 mg/kg. The data indicate that CMDE significantly improved the oral bioavailability of CBD. Overall, CMDE can be used to enhance the oral absorption of poorly bioavailable APIs.
Collapse
Affiliation(s)
- Mounika Aare
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, USA
| | - Arvind Bagde
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, USA
| | - Aakash Nathani
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, USA
| | - Arun K Rishi
- Department of Oncology, Wayne State University, Detroit, MI, USA
| | - Mandip Singh
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, USA.
| |
Collapse
|
6
|
Nunes YC, Mendes NM, Pereira de Lima E, Chehadi AC, Lamas CB, Haber JFS, dos Santos Bueno M, Araújo AC, Catharin VCS, Detregiachi CRP, Laurindo LF, Tanaka M, Barbalho SM, Marin MJS. Curcumin: A Golden Approach to Healthy Aging: A Systematic Review of the Evidence. Nutrients 2024; 16:2721. [PMID: 39203857 PMCID: PMC11357524 DOI: 10.3390/nu16162721] [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: 07/12/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 09/03/2024] Open
Abstract
Aging-related disorders pose significant challenges due to their complex interplay of physiological and metabolic factors, including inflammation, oxidative stress, and mitochondrial dysfunction. Curcumin, a natural compound with potent antioxidant and anti-inflammatory properties, has emerged as a promising candidate for mitigating these age-related processes. However, gaps in understanding the precise mechanisms of curcumin's effects and the optimal dosages for different conditions necessitate further investigation. This systematic review synthesizes current evidence on curcumin's potential in addressing age-related disorders, emphasizing its impact on cognitive function, neurodegeneration, and muscle health in older adults. By evaluating the safety, efficacy, and mechanisms of action of curcumin supplementation, this review aims to provide insights into its therapeutic potential for promoting healthy aging. A systematic search across three databases using specific keywords yielded 2256 documents, leading to the selection of 15 clinical trials for synthesis. Here, we highlight the promising potential of curcumin as a multifaceted therapeutic agent in combating age-related disorders. The findings of this review suggest that curcumin could offer a natural and effective approach to enhancing the quality of life of aging individuals. Further research and well-designed clinical trials are essential to validate these findings and optimize the use of curcumin in personalized medicine approaches for age-related conditions.
Collapse
Affiliation(s)
- Yandra Cervelim Nunes
- Faculdade de Medicina de Marília (FAMEMA), Marília 17519-030, SP, Brazil; (Y.C.N.); (L.F.L.)
| | - Nathalia M. Mendes
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil; (N.M.M.); (E.P.d.L.); (A.C.C.); (J.F.S.H.); (M.d.S.B.); (A.C.A.)
| | - Enzo Pereira de Lima
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil; (N.M.M.); (E.P.d.L.); (A.C.C.); (J.F.S.H.); (M.d.S.B.); (A.C.A.)
| | - Amanda Chabrour Chehadi
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil; (N.M.M.); (E.P.d.L.); (A.C.C.); (J.F.S.H.); (M.d.S.B.); (A.C.A.)
| | - Caroline Barbalho Lamas
- Department of Gerontology, School of Gerontology, Universidade Federal de São Carlos (UFSCar), São Carlos 13565-905, SP, Brazil;
| | - Jesselina F. S. Haber
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil; (N.M.M.); (E.P.d.L.); (A.C.C.); (J.F.S.H.); (M.d.S.B.); (A.C.A.)
| | - Manoela dos Santos Bueno
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil; (N.M.M.); (E.P.d.L.); (A.C.C.); (J.F.S.H.); (M.d.S.B.); (A.C.A.)
| | - Adriano Cressoni Araújo
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil; (N.M.M.); (E.P.d.L.); (A.C.C.); (J.F.S.H.); (M.d.S.B.); (A.C.A.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil; (V.C.S.C.); (C.R.P.D.)
| | - Vitor C. Strozze Catharin
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil; (V.C.S.C.); (C.R.P.D.)
| | - Claudia Rucco P. Detregiachi
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil; (V.C.S.C.); (C.R.P.D.)
| | - Lucas Fornari Laurindo
- Faculdade de Medicina de Marília (FAMEMA), Marília 17519-030, SP, Brazil; (Y.C.N.); (L.F.L.)
| | - Masaru Tanaka
- Danube Neuroscience Research Laboratory, HUN-REN-SZTE Neuroscience Research Group, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Tisza Lajos krt. 113, H-6725 Szeged, Hungary
| | - Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil; (N.M.M.); (E.P.d.L.); (A.C.C.); (J.F.S.H.); (M.d.S.B.); (A.C.A.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil; (V.C.S.C.); (C.R.P.D.)
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Marília 17500-000, SP, Brazil
- Research Coordination, Hospital Beneficente (HBU), University of Marília (UNIMAR), Marília 17525-160, SP, Brazil
| | | |
Collapse
|
7
|
Las Heras K, Garcia-Orue I, Rancan F, Igartua M, Santos-Vizcaino E, Hernandez RM. Modulating the immune system towards a functional chronic wound healing: A biomaterials and Nanomedicine perspective. Adv Drug Deliv Rev 2024; 210:115342. [PMID: 38797316 DOI: 10.1016/j.addr.2024.115342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 05/16/2024] [Accepted: 05/18/2024] [Indexed: 05/29/2024]
Abstract
Chronic non-healing wounds persist as a substantial burden for healthcare systems, influenced by factors such as aging, diabetes, and obesity. In contrast to the traditionally pro-regenerative emphasis of therapies, the recognition of the immune system integral role in wound healing has significantly grown, instigating an approach shift towards immunological processes. Thus, this review explores the wound healing process, highlighting the engagement of the immune system, and delving into the behaviors of innate and adaptive immune cells in chronic wound scenarios. Moreover, the article investigates biomaterial-based strategies for the modulation of the immune system, elucidating how the adjustment of their physicochemical properties or their synergistic combination with other agents such as drugs, proteins or mesenchymal stromal cells can effectively modulate the behaviors of different immune cells. Finally this review explores various strategies based on synthetic and biological nanostructures, including extracellular vesicles, to finely tune the immune system as natural immunomodulators or therapeutic nanocarriers with promising biophysical properties.
Collapse
Affiliation(s)
- Kevin Las Heras
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV-EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Itxaso Garcia-Orue
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV-EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN). Institute of Health Carlos III, Madrid, Spain
| | - Fiorenza Rancan
- Department of Dermatology, Venereology und Allergology,Clinical Research Center for Hair and Skin Science, Charité - Universitätsmedizin Berlin
| | - Manoli Igartua
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV-EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN). Institute of Health Carlos III, Madrid, Spain
| | - Edorta Santos-Vizcaino
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV-EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN). Institute of Health Carlos III, Madrid, Spain.
| | - Rosa Maria Hernandez
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV-EHU), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN). Institute of Health Carlos III, Madrid, Spain.
| |
Collapse
|
8
|
Baruah H, Sarma A, Basak D, Das M. Exosome: From biology to drug delivery. Drug Deliv Transl Res 2024; 14:1480-1516. [PMID: 38252268 DOI: 10.1007/s13346-024-01515-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/27/2023] [Indexed: 01/23/2024]
Abstract
In recent years, different advancements have been observed in nanosized drug delivery systems. Factors such as stability, safety and targeting efficiency cause hindrances in the clinical translation of these synthetic nanocarriers. Therefore, researchers employed endogenous nanocarriers like exosomes as drug delivery vehicles that have an inherent ability to target more efficiently after appropriate functionalization and show higher biocompatibility and less immunogenicity and facilitate penetration through the biological barriers more quickly than the other available carriers. Exosomes are biologically derived lipid bilayer-enclosed nanosized extracellular vesicles (size ranges from 30 to 150 nm) secreted from both prokaryotic and eukaryotic cells and appears significantly in the extracellular space. These EVs (extracellular vesicles) can exist in different sources, including mammals, plants and microorganisms. Different advanced techniques have been introduced for the isolation of exosomes to overcome the existing barriers present with conventional methods. Extensive research on the application of exosomes in therapeutic delivery for treating various diseases related to central nervous system, bone, cancer, skin, etc. has been employed. Several studies are on different stages of clinical trials, and many exosomes patents have been registered.
Collapse
Affiliation(s)
- Himakshi Baruah
- Advanced Drug Delivery Laboratory, Department of Pharmaceutics, School of Pharmaceutical Sciences, Girijananda Chowdhury University, Guwahati, 781017, Assam, India
| | - Anupam Sarma
- Advanced Drug Delivery Laboratory, Department of Pharmaceutics, School of Pharmaceutical Sciences, Girijananda Chowdhury University, Guwahati, 781017, Assam, India.
| | - Debojeet Basak
- Advanced Drug Delivery Laboratory, Department of Pharmaceutics, School of Pharmaceutical Sciences, Girijananda Chowdhury University, Guwahati, 781017, Assam, India
| | - Mridusmita Das
- Advanced Drug Delivery Laboratory, Department of Pharmaceutics, School of Pharmaceutical Sciences, Girijananda Chowdhury University, Guwahati, 781017, Assam, India
| |
Collapse
|
9
|
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
|
10
|
Ran B, Ren X, Lin X, Teng Y, Xin F, Ma W, Zhao X, Li M, Wang J, Wang C, Sun L, Zhang J. Glycyrrhetinic acid loaded in milk-derived extracellular vesicles for inhalation therapy of idiopathic pulmonary fibrosis. J Control Release 2024; 370:811-820. [PMID: 38754632 DOI: 10.1016/j.jconrel.2024.05.024] [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: 03/13/2024] [Revised: 04/27/2024] [Accepted: 05/13/2024] [Indexed: 05/18/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, and life-threatening lung disease for which treatment options are limited. Glycyrrhetinic acid (GA) is a triterpenoid with multiple biological effects, such as anti-inflammatory and anti-fibrotic properties. Herein, inhalable milk-derived extracellular vesicles (mEVs) encapsulating GA (mEVs@GA) were screened and evaluated for IPF treatment. The results indicated that the loading efficiency of GA in mEVs@GA was 8.65%. Therapeutic effects of inhalable mEVs@GA were investigated in vitro and in vivo. The mEVs@GA demonstrated superior anti-inflammatory effects on LPS-stimulated MHS cells. Furthermore, repeated noninvasive inhalation delivery of mEVs@GA in bleomycin-induced IPF mice could decrease the levels of transforming growth factors β1 (TGF-β1), Smad3 and inflammatory cytokines IL-6, IL-1β and TNF-α. The mEVs@GA effectively diminished the development of fibrosis and improved pulmonary function in the IPF mice model at a quarter of the dose compared with the pirfenidone oral administration group. Additionally, compared to pirfenidone-loaded mEVs, mEVs@GA demonstrated superior efficacy at the same drug concentration in the pharmacodynamic study. Overall, inhaled mEVs@GA have the potential to serve as an effective therapeutic option in the treatment of IPF.
Collapse
Affiliation(s)
- Bo Ran
- Shenyang Pharmaceutiacal University, Shenyang 110016, China; Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; Yangtze Delta Drug Advanced Research Institute, Nantong 226126, China
| | - Xiaohong Ren
- Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Xueyuan Lin
- Shenyang Pharmaceutiacal University, Shenyang 110016, China; Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; Yangtze Delta Drug Advanced Research Institute, Nantong 226126, China
| | - Yupu Teng
- Shenyang Pharmaceutiacal University, Shenyang 110016, China; Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; Yangtze Delta Drug Advanced Research Institute, Nantong 226126, China
| | - Fangyuan Xin
- Shenyang Pharmaceutiacal University, Shenyang 110016, China; Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; Yangtze Delta Drug Advanced Research Institute, Nantong 226126, China
| | - Wuzhen Ma
- Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiangyu Zhao
- Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; Yangtze Delta Drug Advanced Research Institute, Nantong 226126, China
| | - Mingwei Li
- Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; Yangtze Delta Drug Advanced Research Institute, Nantong 226126, China
| | - Jinghuang Wang
- Shenyang Pharmaceutiacal University, Shenyang 110016, China; Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; Yangtze Delta Drug Advanced Research Institute, Nantong 226126, China
| | - Caifen Wang
- Shenyang Pharmaceutiacal University, Shenyang 110016, China; Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China
| | - Lixin Sun
- Shenyang Pharmaceutiacal University, Shenyang 110016, China.
| | - Jiwen Zhang
- Shenyang Pharmaceutiacal University, Shenyang 110016, China; Center for Drug Delivery System, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China; Yangtze Delta Drug Advanced Research Institute, Nantong 226126, China.
| |
Collapse
|
11
|
Jiang Q, Liu Y, Si X, Wang L, Gui H, Tian J, Cui H, Jiang H, Dong W, Li B. Potential of Milk-Derived Extracellular Vesicles as Carriers for Oral Delivery of Active Phytoconstituents. Annu Rev Food Sci Technol 2024; 15:431-454. [PMID: 38359948 DOI: 10.1146/annurev-food-072023-034354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Extracellular vesicles (EVs) play a crucial role in intercellular communication and have the potential to serve as in vivo carriers for delivering active molecules. The biocompatibility advantages of EVs over artificial nanocarriers create new frontiers for delivering modern active molecules. Milk is a favorable source of EVs because of its high bioavailability, low immunogenicity, and commercial producibility. In this review, we analyzed the advantages of milk-derived EVs in the oral delivery of active molecules, discussed their research progress in delivering active phytoconstituents, and summarized the necessary technologies and critical unit operations required for the development of an oral delivery system based on EVs. The review aims to provide innovative ideas and fundamental quality control guidelines for developing the next-generation oral drug delivery system based on milk-derived EVs.
Collapse
Affiliation(s)
- Qiao Jiang
- College of Food Science, Shenyang Agricultural University, Shenyang, China;
| | - Yubo Liu
- College of Food Science, Shenyang Agricultural University, Shenyang, China;
| | - Xu Si
- College of Food Science, Shenyang Agricultural University, Shenyang, China;
| | - Li Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Hailong Gui
- College of Food Science, Shenyang Agricultural University, Shenyang, China;
| | - Jinlong Tian
- College of Food Science, Shenyang Agricultural University, Shenyang, China;
| | - Huijun Cui
- College of Food Science, Shenyang Agricultural University, Shenyang, China;
| | - Hongzhou Jiang
- Anhui Ziyue Biological Technology Co., Ltd., Wuhu, China
| | - Wenjiang Dong
- Spice and Beverage Research Institute, Chinese Academy of Tropical Agricultural Sciences, Wanning, China
| | - Bin Li
- College of Food Science, Shenyang Agricultural University, Shenyang, China;
| |
Collapse
|
12
|
Jin Z, Na J, Lin X, Jiao R, Liu X, Huang Y. Plant-derived exosome-like nanovesicles: A novel nanotool for disease therapy. Heliyon 2024; 10:e30630. [PMID: 38765146 PMCID: PMC11098843 DOI: 10.1016/j.heliyon.2024.e30630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 04/27/2024] [Accepted: 05/01/2024] [Indexed: 05/21/2024] Open
Abstract
Exosomes are extracellular vesicles comprising bilayer phospholipid membranes and are secreted by eukaryotic cells. They are released via cellular exocytosis, contain DNA, RNA, proteins, and other substances, and participate in various cellular communications between tissues and organs. Since the discovery of exosomes in 1983, animal-derived exosomes have become a research focus for small-molecule drug delivery in biology, medicine, and other fields owing to their good biocompatibility and homing effects. Recent studies have found that plant-derived exosome-like nanovesicles (PELNVs) exhibit certain biological effects, such as anti-inflammatory and anti-tumor abilities, and have minimal toxic side effects. Because they are rich in active lipid molecules with certain pharmacological effects, PELNVs could be novel carriers for drug delivery. In this review, the biological formation and effects, isolation, and extraction of PELNVs, as well as characteristics of transporting drugs as carriers are summarized to provide new ideas and methods for future research on plant-derived exosome-like nanovesicles.
Collapse
Affiliation(s)
- Ze Jin
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Jintong Na
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Xia Lin
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Rong Jiao
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Xiyu Liu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Yong Huang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| |
Collapse
|
13
|
Hillman T. The application of plant-exosome-like nanovesicles as improved drug delivery systems for cancer vaccines. Discov Oncol 2024; 15:136. [PMID: 38683256 PMCID: PMC11058161 DOI: 10.1007/s12672-024-00974-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 04/08/2024] [Indexed: 05/01/2024] Open
Abstract
The use of cancer immunotherapeutics is currently increasing. Cancer vaccines, as a form of immunotherapy, are gaining much attention in the medical community since specific tumor-antigens can activate immune cells to induce an anti-tumor immune response. However, the delivery of cancer vaccines presents many issues for research scientists when designing cancer treatments and requires further investigation. Nanoparticles, synthetic liposomes, bacterial vectors, viral particles, and mammalian exosomes have delivered cancer vaccines. In contrast, the use of many of these nanotechnologies produces many issues of cytotoxicity, immunogenicity, and rapid clearance by the mononuclear phagocyte system (MPS). Plant-exosome-like nanovesicles (PELNVs) can provide solutions for many of these challenges because they are innocuous and nonimmunogenic when delivering nanomedicines. Hence, this review will describe the potential use of PELNVs to deliver cancer vaccines. In this review, different approaches of cancer vaccine delivery will be detailed, the mechanism of oral vaccination for delivering cancer vaccines will be described, and the review will discuss the use of PELNVs as improved drug delivery systems for cancer vaccines via oral administration while also addressing the subsequent challenges for advancing their usage into the clinical setting.
Collapse
|
14
|
Salehi M, Negahdari B, Mehryab F, Shekari F. Milk-Derived Extracellular Vesicles: Biomedical Applications, Current Challenges, and Future Perspectives. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:8304-8331. [PMID: 38587896 DOI: 10.1021/acs.jafc.3c07899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Extracellular vesicles (EVs) are nano to-micrometer-sized sacs that are released by almost all animal and plant cells and act as intercellular communicators by transferring their cargos between the source and target cells. As a safe and scalable alternative to conditioned medium-derived EVs, milk-derived EVs (miEVs) have recently gained a great deal of popularity. Numerous studies have shown that miEVs have intrinsic therapeutic actions that can treat diseases and enhance human health. Additionally, they can be used as natural drug carriers and novel classes of biomarkers. However, due to the complexity of the milk, the successful translation of miEVs from benchtop to bedside still faces several unfilled gaps, especially a lack of standardized protocols for the isolation of high-purity miEVs. In this work, by comprehensively reviewing the bovine miEVs studies, we provide an overview of current knowledge and research on miEVs while highlighting their challenges and enormous promise as a novel class of theranostics. It is hoped that this study will pave the way for clinical applications of miEVs by addressing their challenges and opportunities.
Collapse
Affiliation(s)
- Mahsa Salehi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran 14177-55469, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148, Iran
| | - Babak Negahdari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran 14177-55469, Iran
| | - Fatemeh Mehryab
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148, Iran
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran 14155-6153, Iran
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148, Iran
- Advanced Therapy Medicinal Product Technology Development Center (ATMP-TDC), Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 16635-148, Iran
| |
Collapse
|
15
|
Dai C, Xu Q, Li L, Liu Y, Qu S. Milk Extracellular Vesicles: Natural Nanoparticles for Enhancing Oral Drug Delivery against Bacterial Infections. ACS Biomater Sci Eng 2024; 10:1988-2000. [PMID: 38529792 DOI: 10.1021/acsbiomaterials.3c01824] [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: 03/27/2024]
Abstract
Oral drug delivery is typically preferred as a therapeutic intervention due to the complexities and expenses associated with intravenous administration. However, some drugs are poorly absorbed orally, requiring intravenous administration to bypass the gastrointestinal tract and deliver the drug directly into the bloodstream. Thus, there is an urgent need to develop novel drug delivery platforms to overcome the challenges of oral drug delivery with low solubility, low permeability, oral degradation, and low bioavailability. Advances in extracellular vesicles (EVs) as natural carriers have provided emerging approaches to improve potential therapeutic applications. Milk not only contains traditional nutrients but is also rich in EVs. In this Review, we focus mainly on the purification of milk EVs (mEVs), their safety, and the advantages of mEV-based drug carriers in combatting intestinal infections. Additionally, we summarize several advantages of mEVs over conventional synthetic carriers, such as low immunogenicity, high biocompatibility, and the ability to transfer bioactive molecules between cells. Considering the unmet gaps of mEVs in clinical translation, it is essential to review the cargo loading into mEVs and future perspectives for their use as natural drug carriers for oral delivery. This overview of mEV-based drug carriers for oral delivery sheds light on alternative approaches to treat clinical infections associated with intestinal pathogens and the development of novel oral delivery systems.
Collapse
Affiliation(s)
- Cunchun Dai
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Qingjun Xu
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Lin Li
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Ying Liu
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China
| | - Shaoqi Qu
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| |
Collapse
|
16
|
Albaladejo-García V, Morán L, Santos-Coquillat A, González MI, Ye H, Vázquez Ogando E, Vaquero J, Cubero FJ, Desco M, Salinas B. Curcumin encapsulated in milk small extracellular vesicles as a nanotherapeutic alternative in experimental chronic liver disease. Biomed Pharmacother 2024; 173:116381. [PMID: 38452655 DOI: 10.1016/j.biopha.2024.116381] [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: 11/07/2023] [Revised: 02/27/2024] [Accepted: 02/29/2024] [Indexed: 03/09/2024] Open
Abstract
Curcumin is a natural molecule widely tested in preclinical and clinical studies due to its antioxidant and anti-inflammatory activity. Nevertheless, its high hydrophobicity and low bioavailability limit in vivo applications. To overcome curcumin´s drawbacks, small extracellular vesicles (sEVs) have emerged as potential drug delivery systems due to their non-immunogenicity, nanometric size and amphiphilic composition. This work presents curcumin cargo into milk sEV structure and further in vitro and in vivo evaluation as a therapeutic nanoplatform. The encapsulation of curcumin into sEV was performed by two methodologies under physiological conditions: a passive incorporation and active cargo employing saponin. Loaded sEVs (sEVCurPas and sEVCurAc) were fully characterized by physicochemical techniques, confirming that neither methodology affects the morphology or size of the nanoparticles (sEV: 113.3±5.1 nm, sEVCurPas: 127.0±4.5 nm and sEVCurAc: 98.5±3.6 nm). Through the active approach with saponin (sEVCurAc), a three-fold higher cargo was obtained (433.5 µg/mL) in comparison with the passive approach (129.1 µg/mL). These sEVCurAc were further evaluated in vitro by metabolic activity assay (MTT), confocal microscopy, and flow cytometry, showing a higher cytotoxic effect in the tumoral cells RAW264.7 and HepG2 than in primary hepatocytes, specially at high doses of sEVCurAc (4%, 15% and 30% of viability). In vivo evaluation in an experimental model of liver fibrosis confirmed sEVCurAc therapeutic effects, leading to a significant decrease of serum markers of liver damage (ALT) (557 U/L to 338 U/L with sEVCurAc therapy) and a tendency towards decreased liver fibrogenesis and extracellular matrix (ECM) deposition.
Collapse
Affiliation(s)
- Virginia Albaladejo-García
- Unidad de Medicina y Cirugía Experimental, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid 28007, Spain
| | - Laura Morán
- Departamento de Inmunología, Oftalmología y ENT, Facultad de Medicina de la Universidad Complutense de Madrid, Madrid 28040, Spain
| | - Ana Santos-Coquillat
- Unidad de Medicina y Cirugía Experimental, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid 28007, Spain; Unidad de Imagen Avanzada, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
| | - María I González
- Unidad de Medicina y Cirugía Experimental, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid 28007, Spain; Unidad de Imagen Avanzada, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
| | - Hui Ye
- Departamento de Inmunología, Oftalmología y ENT, Facultad de Medicina de la Universidad Complutense de Madrid, Madrid 28040, Spain
| | - Elena Vázquez Ogando
- HepatoGastro Lab, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid 28007, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Javier Vaquero
- HepatoGastro Lab, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid 28007, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Francisco Javier Cubero
- Departamento de Inmunología, Oftalmología y ENT, Facultad de Medicina de la Universidad Complutense de Madrid, Madrid 28040, Spain; HepatoGastro Lab, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid 28007, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Manuel Desco
- Unidad de Medicina y Cirugía Experimental, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid 28007, Spain; Unidad de Imagen Avanzada, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain; Departamento de Bioingeniería, Universidad Carlos III de Madrid, Madrid 28911, Spain; CIBER de Salud Mental, Instituto de Salud Carlos III, Madrid, Spain.
| | - Beatriz Salinas
- Unidad de Medicina y Cirugía Experimental, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid 28007, Spain; Unidad de Imagen Avanzada, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain; Departamento de Bioingeniería, Universidad Carlos III de Madrid, Madrid 28911, Spain; CIBER de Salud Mental, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
17
|
Han G, Kim H, Jang H, Kim ES, Kim SH, Yang Y. Oral TNF-α siRNA delivery via milk-derived exosomes for effective treatment of inflammatory bowel disease. Bioact Mater 2024; 34:138-149. [PMID: 38223538 PMCID: PMC10784143 DOI: 10.1016/j.bioactmat.2023.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/05/2023] [Accepted: 12/12/2023] [Indexed: 01/16/2024] Open
Abstract
Oral administration facilitates the direct delivery of drugs to lesions within the small intestine and colon, making it an ideal approach for treating patients with inflammatory bowel disease. However, multiple physical barriers impede the delivery of oral RNA drugs through the gastrointestinal tract. Herein, we developed a novel oral siRNA delivery system that protects nucleic acids in extreme environments by employing exosomes derived from milk to encapsulate tumor necrosis factor-alpha (TNF-α) siRNA completely. The remarkable structural stability of milk-derived exosomes (M-Exos), as opposed to those from HEK293T cells, makes them exceptional siRNA carriers. Results demonstrate that milk exosomes loaded with TNF-α siRNA (M-Exo/siR) can effectively inhibit the expression of TNF-α-related inflammatory cytokines. Moreover, given that milk exosomes are composed of unique lipids with high bioavailability, orally administered M-Exo/siR effectively reach colonic tissues, leading to decreased TNF-α expression and successful alleviation of colitis symptoms in a dextran sulfate sodium-induced inflammatory bowel disease murine model. Hence, milk-derived exosomes carrying TNF-α siRNA can be effectively employed to treat inflammatory bowel disease. Indeed, using exosomes naturally derived from milk may shift the current paradigm of oral gene delivery, including siRNA.
Collapse
Affiliation(s)
- Geonhee Han
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Hyosuk Kim
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Hochung Jang
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology, Seoul, 02792, Republic of Korea
| | - Eun Sun Kim
- Department of Internal Medicine, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Sun Hwa Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Yoosoo Yang
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology, Seoul, 02792, Republic of Korea
| |
Collapse
|
18
|
Zhang W, Song M, Fang Z, Chen F, Yuan H, Gao X, Liu K. Role of extracellular vesicles in insulin resistance: Signaling pathways, bioactive substances, miRNAs, and therapeutic potential. Cell Biochem Funct 2024; 42:e4013. [PMID: 38639198 DOI: 10.1002/cbf.4013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 03/31/2024] [Accepted: 04/03/2024] [Indexed: 04/20/2024]
Abstract
Extracellular vesicles are small lipid bilayer particles that resemble the structure of cells and range in size from 30 to 1000 nm. They transport a variety of physiologically active molecules, such as proteins, lipids, and miRNAs. Insulin resistance (IR) is a pathological disease in which insulin-responsive organs or components become less sensitive to insulin's physiological effects, resulting in decreased glucose metabolism in target organs such as the liver, muscle, and adipose tissue. Extracellular vesicles have received a lot of attention as essential intercellular communication mediators in the setting of IR. This review looks at extracellular vesicles' role in IR from three angles: signaling pathways, bioactive compounds, and miRNAs. Relevant publications are gathered to investigate the induction, inhibition, and bidirectional regulation of extracellular vesicles in IR, as well as their role in insulin-related illnesses. Furthermore, considering the critical function of extracellular vesicles in regulating IR, the study analyzes the practicality of employing extracellular vesicles for medication delivery and the promise of combination therapy for IR.
Collapse
Affiliation(s)
- Wang Zhang
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Mengdi Song
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Zhou Fang
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Feng Chen
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Hui Yuan
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Xinran Gao
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Kehai Liu
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
- Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai, China
| |
Collapse
|
19
|
Zeng B, Li Y, Xia J, Xiao Y, Khan N, Jiang B, Liang Y, Duan L. Micro Trojan horses: Engineering extracellular vesicles crossing biological barriers for drug delivery. Bioeng Transl Med 2024; 9:e10623. [PMID: 38435823 PMCID: PMC10905561 DOI: 10.1002/btm2.10623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 10/05/2023] [Accepted: 11/09/2023] [Indexed: 03/05/2024] Open
Abstract
The biological barriers of the body, such as the blood-brain, placental, intestinal, skin, and air-blood, protect against invading viruses and bacteria while providing necessary physical support. However, these barriers also hinder the delivery of drugs to target tissues, reducing their therapeutic efficacy. Extracellular vesicles (EVs), nanostructures with a diameter ranging from 30 nm to 10 μm secreted by cells, offer a potential solution to this challenge. These natural vesicles can effectively pass through various biological barriers, facilitating intercellular communication. As a result, artificially engineered EVs that mimic or are superior to the natural ones have emerged as a promising drug delivery vehicle, capable of delivering drugs to almost any body part to treat various diseases. This review first provides an overview of the formation and cross-species uptake of natural EVs from different organisms, including animals, plants, and bacteria. Later, it explores the current clinical applications, perspectives, and challenges associated with using engineered EVs as a drug delivery platform. Finally, it aims to inspire further research to help bioengineered EVs effectively cross biological barriers to treat diseases.
Collapse
Affiliation(s)
- Bin Zeng
- Graduate SchoolGuangxi University of Chinese MedicineNanningGuangxiChina
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Artificial Intelligence Biomedical Innovation Platform, Shenzhen Second People's Hospitalthe First Affiliated Hospital of Shenzhen UniversityShenzhenGuangdongChina
| | - Ying Li
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Artificial Intelligence Biomedical Innovation Platform, Shenzhen Second People's Hospitalthe First Affiliated Hospital of Shenzhen UniversityShenzhenGuangdongChina
| | - Jiang Xia
- Department of ChemistryThe Chinese University of Hong Kong, ShatinHong Kong SARChina
| | - Yin Xiao
- School of Medicine and Dentistry & Menzies Health Institute Queensland, SouthportGold CoastQueenslandAustralia
| | - Nawaz Khan
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Artificial Intelligence Biomedical Innovation Platform, Shenzhen Second People's Hospitalthe First Affiliated Hospital of Shenzhen UniversityShenzhenGuangdongChina
| | - Bin Jiang
- Graduate SchoolGuangxi University of Chinese MedicineNanningGuangxiChina
- R&D Division, Eureka Biotech Inc, PhiladelphiaPennsylvaniaUSA
| | - Yujie Liang
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning HospitalShenzhen Mental Health Center, Shenzhen Key Laboratory for Psychological Healthcare and Shenzhen Institute of Mental HealthShenzhenGuangdongChina
| | - Li Duan
- Graduate SchoolGuangxi University of Chinese MedicineNanningGuangxiChina
- Department of Orthopedics, Shenzhen Intelligent Orthopaedics and Biomedical Innovation Platform, Guangdong Artificial Intelligence Biomedical Innovation Platform, Shenzhen Second People's Hospitalthe First Affiliated Hospital of Shenzhen UniversityShenzhenGuangdongChina
| |
Collapse
|
20
|
Lo KJ, Wang MH, Ho CT, Pan MH. Plant-Derived Extracellular Vesicles: A New Revolutionization of Modern Healthy Diets and Biomedical Applications. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:2853-2878. [PMID: 38300835 DOI: 10.1021/acs.jafc.3c06867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
Plant-derived extracellular vesicles (PDEVs) have recently emerged as a promising area of research due to their potential health benefits and biomedical applications. Produced by various plant species, these EVs contain diverse bioactive molecules, including proteins, lipids, and nucleic acids. Increasing in vitro and in vivo studies have shown that PDEVs have inherent pharmacological activities that affect cellular processes, exerting anti-inflammatory, antioxidant, and anticancer activities, which can potentially contribute to disease therapy and improve human health. Additionally, PDEVs have shown potential as efficient and biocompatible drug delivery vehicles in treating various diseases. However, while PDEVs serve as a potential rising star in modern healthy diets and biomedical applications, further research is needed to address their underlying knowledge gaps, especially the lack of standardized protocols for their isolation, identification, and large-scale production. Furthermore, the safety and efficacy of PDEVs in clinical applications must be thoroughly evaluated. In this review, we concisely discuss current knowledge in the PDEV field, including their characteristics, biomedical applications, and isolation methods, to provide an overview of the current state of PDEV research. Finally, we discuss the challenges regarding the current and prospective issues for PDEVs. This review is expected to provide new insights into healthy diets and biomedical applications of vegetables and fruits, inspiring new advances in natural food-based science and technology.
Collapse
Affiliation(s)
- Kai-Jiun Lo
- Institute of Food Science and Technology, National Taiwan University, Taipei 10617, Taiwan
| | - Mu-Hui Wang
- Department of Medical Research, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, New Jersey 08901-8520, United States
| | - Min-Hsiung Pan
- Institute of Food Science and Technology, National Taiwan University, Taipei 10617, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan
- Department of Health and Nutrition Biotechnology, Asia University, Taichung 41354, Taiwan
| |
Collapse
|
21
|
Ming‐Kun C, Zi‐Xian C, Mao‐Ping C, Hong C, Zhuang‐Fei C, Shan‐Chao Z. Engineered extracellular vesicles: A new approach for targeted therapy of tumors and overcoming drug resistance. Cancer Commun (Lond) 2024; 44:205-225. [PMID: 38155418 PMCID: PMC10876209 DOI: 10.1002/cac2.12518] [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: 08/16/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 12/30/2023] Open
Abstract
Targeted delivery of anti-tumor drugs and overcoming drug resistance in malignant tumor cells remain significant clinical challenges. However, there are only few effective methods to address these issues. Extracellular vesicles (EVs), actively secreted by cells, play a crucial role in intercellular information transmission and cargo transportation. Recent studies have demonstrated that engineered EVs can serve as drug delivery carriers and showed promising application prospects. Nevertheless, there is an urgent need for further improvements in the isolation and purification of EVs, surface modification techniques, drug assembly processes, and precise recognition of tumor cells for targeted drug delivery purposes. In this review, we summarize the applications of engineered EVs in cancer treatment and overcoming drug resistance, and current challenges associated with engineered EVs are also discussed. This review aims to provide new insights and potential directions for utilizing engineered EVs as targeted delivery systems for anti-tumor drugs and overcoming drug resistance in the near future.
Collapse
Affiliation(s)
- Chen Ming‐Kun
- Department of UrologyThe Third Affiliated Hospital of Southern Medical UniversityGuangzhouGuangdongP. R. China
- The Third Clinical CollegeSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Chen Zi‐Xian
- Department of UrologyThe Third Affiliated Hospital of Southern Medical UniversityGuangzhouGuangdongP. R. China
- The Third Clinical CollegeSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Cai Mao‐Ping
- Department of UrologyThe Third Affiliated Hospital of Southern Medical UniversityGuangzhouGuangdongP. R. China
- The Third Clinical CollegeSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Chen Hong
- Luoyang Key Laboratory of Organic Functional MoleculesCollege of Food and DrugLuoyang Normal UniversityLuoyangHenanP. R. China
| | - Chen Zhuang‐Fei
- Department of UrologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| | - Zhao Shan‐Chao
- Department of UrologyThe Third Affiliated Hospital of Southern Medical UniversityGuangzhouGuangdongP. R. China
- The Third Clinical CollegeSouthern Medical UniversityGuangzhouGuangdongP. R. China
- Department of UrologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdongP. R. China
| |
Collapse
|
22
|
Goudarzi F, Jajarmi V, Shojaee S, Mohebali M, Keshavarz H. Formulation and evaluation of atovaquone-loaded macrophage-derived exosomes against Toxoplasma gondii: in vitro and in vivo assessment. Microbiol Spectr 2024; 12:e0308023. [PMID: 38014940 PMCID: PMC10782982 DOI: 10.1128/spectrum.03080-23] [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: 08/11/2023] [Accepted: 10/03/2023] [Indexed: 11/29/2023] Open
Abstract
IMPORTANCE This study is the first of its kind that suggests exosomes as a nano-carrier loaded with atovaquone (ATQ), which could be considered as a new strategy for improving the effectiveness of ATQ against acute and chronic phases of Toxoplasma gondii.
Collapse
Affiliation(s)
- Fatemeh Goudarzi
- Department of Medical Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahid Jajarmi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeedeh Shojaee
- Department of Medical Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Mohebali
- Department of Medical Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Center for Research of Endemic Parasites of Iran (CREPI), Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Keshavarz
- Department of Medical Parasitology and Mycology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Center for Research of Endemic Parasites of Iran (CREPI), Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
23
|
Mondal J, Pillarisetti S, Junnuthula V, Surwase SS, Hwang SR, Park IK, Lee YK. Extracellular vesicles and exosome-like nanovesicles as pioneering oral drug delivery systems. Front Bioeng Biotechnol 2024; 11:1307878. [PMID: 38260737 PMCID: PMC10800420 DOI: 10.3389/fbioe.2023.1307878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 12/15/2023] [Indexed: 01/24/2024] Open
Abstract
As extracellular vesicle (EV)-based nanotechnology has developed rapidly, it has made unprecedented opportunities for nanomedicine possible. EVs and exosome-like nanovesicles (ELNVs) are natural nanocarriers with unique structural, compositional, and morphological characteristics that provide excellent physical, chemical, and biochemical properties. In this literature, we examine the characteristics of EVs, including how they are administered orally and their therapeutic activity. According to the current examples of EVs and ELNVs for oral delivery, milk and plant EVs can exert therapeutic effects through their protein, nucleic acid, and lipid components. Furthermore, several methods for loading drugs into exosomes and targeting exosomes have been employed to investigate their therapeutic capability. Moreover, we discuss EVs as potential drug carriers and the potential role of ELNVs for disease prevention and treatment or as potential drug carriers in the future. In conclusion, the issues associated with the development of EVs and ELNVs from sources such as milk and plants, as well as concerns with standardized applications of these EVs, are discussed.
Collapse
Affiliation(s)
- Jagannath Mondal
- Department of Green Bioengineering, Korea National University of Transportation, Chungju, Republic of Korea
| | - Shameer Pillarisetti
- Department of Biomedical Sciences and Biomedical Science Graduate Program (BMSGP), Chonnam National University Medical School, Gwangju, Republic of Korea
| | | | - Sachin S. Surwase
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Seung Rim Hwang
- College of Pharmacy, Chosun University, Gwangju, Republic of Korea
| | - In-Kyu Park
- Department of Biomedical Sciences and Biomedical Science Graduate Program (BMSGP), Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Yong-kyu Lee
- Department of Green Bioengineering, Korea National University of Transportation, Chungju, Republic of Korea
- Department of Chemical and Biological Engineering, Korea National University of Transportation, Chungju, Republic of Korea
| |
Collapse
|
24
|
Afzal A, Khawar MB, Habiba U, Afzal H, Hamid SE, Rafiq M, Abbasi MH, Sheikh N, Abaidullah R, Asif Z, Saeed T. Diagnostic and therapeutic value of EVs in lungs diseases and inflammation. Mol Biol Rep 2023; 51:26. [PMID: 38127201 DOI: 10.1007/s11033-023-09045-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 11/02/2023] [Indexed: 12/23/2023]
Abstract
Extracellular vesicles (EVs) are membrane-derived messengers which have been playing an important role in the inflammation and pathogenesis of lung diseases. EVs contain varieties of DNA, RNA, and membrane receptors through which they work as a delivery system for bioactive molecules as well as intracellular communicators. EV signaling mediates tumor progression and metastasis. EVs are linked with many diseases and perform a diagnostic role in lung injury and inflammation so are used to diagnose the severity of diseases. EVs containing a variety of biomolecules communicate with the recipient cells during pathophysiological mechanisms thereby acquiring the attention of clinicians toward the diagnostic and therapeutic potential of EVs in different lung diseases. In this review, we summarize the role of EVs in inflammation with an emphasis on their potential as a novel candidate in the diagnostics and therapeutics of chronic obstructive pulmonary disease, asthma, and sarcoidosis.
Collapse
Affiliation(s)
- Ali Afzal
- Molecular Medicine and Cancer Therapeutics Lab, Department of Zoology, Faculty of Sciences, University of Central Punjab, Lahore, Pakistan
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Muhammad Babar Khawar
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China.
- Applied Molecular Biology and Biomedicine Lab, Department of Zoology, University of Narowal, Narowal, Pakistan.
| | - Ume Habiba
- Department of Zoology, University of Education, Lahore, Pakistan
| | - Hanan Afzal
- Molecular Medicine and Cancer Therapeutics Lab, Department of Zoology, Faculty of Sciences, University of Central Punjab, Lahore, Pakistan
| | - Syeda Eisha Hamid
- Molecular Medicine and Cancer Therapeutics Lab, Department of Zoology, Faculty of Sciences, University of Central Punjab, Lahore, Pakistan
| | - Mussarat Rafiq
- Cell & Molecular Biology Lab, Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | | | - Nadeem Sheikh
- Cell & Molecular Biology Lab, Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | - Rimsha Abaidullah
- Applied Molecular Biology and Biomedicine Lab, Department of Zoology, University of Narowal, Narowal, Pakistan
| | - Zoya Asif
- Applied Molecular Biology and Biomedicine Lab, Department of Zoology, University of Narowal, Narowal, Pakistan
| | - Tahaa Saeed
- Department of Biology, Syed Babar Ali School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| |
Collapse
|
25
|
Cui Z, Amevor FK, Zhao X, Mou C, Pang J, Peng X, Liu A, Lan X, Liu L. Potential therapeutic effects of milk-derived exosomes on intestinal diseases. J Nanobiotechnology 2023; 21:496. [PMID: 38115131 PMCID: PMC10731872 DOI: 10.1186/s12951-023-02176-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/25/2023] [Indexed: 12/21/2023] Open
Abstract
Exosomes are extracellular vesicles with the diameter of 30 ~ 150 nm, and are widely involved in intercellular communication, disease diagnosis and drug delivery carriers for targeted disease therapy. Therapeutic application of exosomes as drug carriers is limited due to the lack of sources and methods for obtaining adequate exosomes. Milk contains abundant exosomes, several studies have shown that milk-derived exosomes play crucial roles in preventing and treating intestinal diseases. In this review, we summarized the biogenesis, secretion and structure, current novel methods used for the extraction and identification of exosomes, as well as discussed the role of milk-derived exosomes in treating intestinal diseases, such as inflammatory bowel disease, necrotizing enterocolitis, colorectal cancer, and intestinal ischemia and reperfusion injury by regulating intestinal immune homeostasis, restoring gut microbiota composition and improving intestinal structure and integrity, alleviating conditions such as oxidative stress, cell apoptosis and inflammation, and reducing mitochondrial reactive oxygen species (ROS) and lysosome accumulation in both humans and animals. In addition, we discussed future prospects for the standardization of milk exosome production platform to obtain higher concentration and purity, and complete exosomes derived from milk. Several in vivo clinical studies are needed to establish milk-derived exosomes as an effective and efficient drug delivery system, and promote its application in the treatment of various diseases in both humans and animals.
Collapse
Affiliation(s)
- Zhifu Cui
- College of Animal Science and Technology, Southwest University, Chongqing, P. R. China
| | - Felix Kwame Amevor
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Sichuan, P. R. China
| | - Xingtao Zhao
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Sichuan, P. R. China
| | - Chunyan Mou
- College of Animal Science and Technology, Southwest University, Chongqing, P. R. China
| | - Jiaman Pang
- College of Animal Science and Technology, Southwest University, Chongqing, P. R. China
| | - Xie Peng
- College of Animal Science and Technology, Southwest University, Chongqing, P. R. China
| | - Anfang Liu
- College of Animal Science and Technology, Southwest University, Chongqing, P. R. China
| | - Xi Lan
- College of Animal Science and Technology, Southwest University, Chongqing, P. R. China.
| | - Lingbin Liu
- College of Animal Science and Technology, Southwest University, Chongqing, P. R. China.
- College of Animal Science and Technology, Chongqing Key Laboratory of Forage & Herbivore, Chongqing Engineering Research Center for Herbivores Resource Protection and Utilization, Southwest University, Beibei, Chongqing, 400715, P. R. China.
| |
Collapse
|
26
|
Hur JY, Lee S, Shin WR, Kim YH, Ahn JY. The emerging role of medical foods and therapeutic potential of medical food-derived exosomes. NANOSCALE ADVANCES 2023; 6:32-50. [PMID: 38125597 PMCID: PMC10729880 DOI: 10.1039/d3na00649b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 11/08/2023] [Indexed: 12/23/2023]
Abstract
Medical food is consumed for the purpose of improving specific nutritional requirements or disease conditions, such as inflammation, diabetes, and cancer. It involves partial or exclusive feeding for fulfilling unique nutritional requirements of patients and is different from medicine, consisting of basic nutrients, such as polyphenols, vitamins, sugars, proteins, lipids, and other functional ingredients to nourish the patients. Recently, studies on extracellular vesicles (exosomes) with therapeutic and drug carrier potential have been actively conducted. In addition, there have been attempts to utilize exosomes as medical food components. Consequently, the application of exosomes is expanding in different fields with increasing research being conducted on their stability and safety. Herein, we introduced the current trends of medical food and the potential utilization of exosomes in them. Moreover, we proposed Medi-Exo, a exosome-based medical food. Furthermore, we comprehensively elucidate various disease aspects between medical food-derived exosomes (Medi-Exo) and therapeutic natural bionanocomposites. This review highlights the therapeutic challenges regarding Medi-Exo and its potential health benefits.
Collapse
Affiliation(s)
- Jin-Young Hur
- Department of Microbiology, Chungbuk National University 1 Chungdae-Ro, Seowon-Gu Cheongju 28644 South Korea +82-43-264-9600 +82-43-261-2301 +82-43-261-3575
| | - SeonHyung Lee
- Department of Bioengineering, University of Pennsylvania 210 S 33rd St. Philadelphia PA 19104 USA
| | - Woo-Ri Shin
- Department of Microbiology, Chungbuk National University 1 Chungdae-Ro, Seowon-Gu Cheongju 28644 South Korea +82-43-264-9600 +82-43-261-2301 +82-43-261-3575
- Department of Bioengineering, University of Pennsylvania 210 S 33rd St. Philadelphia PA 19104 USA
| | - Yang-Hoon Kim
- Department of Microbiology, Chungbuk National University 1 Chungdae-Ro, Seowon-Gu Cheongju 28644 South Korea +82-43-264-9600 +82-43-261-2301 +82-43-261-3575
| | - Ji-Young Ahn
- Department of Microbiology, Chungbuk National University 1 Chungdae-Ro, Seowon-Gu Cheongju 28644 South Korea +82-43-264-9600 +82-43-261-2301 +82-43-261-3575
| |
Collapse
|
27
|
Yu X, Dong M, Wang L, Yang Q, Wang L, Han W, Dong J, Liu T, Kong Y, Niu W. Nanotherapy for bone repair: milk-derived small extracellular vesicles delivery of icariin. Drug Deliv 2023; 30:2169414. [PMID: 36714914 PMCID: PMC9888478 DOI: 10.1080/10717544.2023.2169414] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Icariin (ICA) played an important role in the treatment of inflammatory bone defects. However, pharmacokinetic studies have shown that its poor bioavailability limited its application. In this context, we isolated bovine milk-derived sEV and prepared sEV-ICA to improve the osteogenic effect of ICA. In this study, we successfully constructed sEV-ICA. sEV-ICA was found to have significantly higher osteogenic efficiency than ICA in cell culture and cranial bone defect models. Mechanistically, bioinformatics analysis predicted that signal transducers and activators of transcription 5 (STAT5a) may bind to the GJA1 promoter, while luciferase activity assays and chromatin immunoprecipitation (ChIP) experiments confirmed that STAT5a directly binded to the GJA1 promoter to promote osteogenesis. We proved that compared with ICA, sEV-ICA showed a better effect of promoting bone repair in vivo and in vitro. In addition, sEV-ICA could promote osteogenesis by promoting the combination of STAT5a and GJA1 promoter. In summary, as a complex drug delivery system, sEV-ICA constituted a rapid and effective method for the treatment of bone defects and could improve the osteogenic activity of ICA.
Collapse
Affiliation(s)
- Xinxin Yu
- School of Stomatology, Dalian Medical University, Dalian, Liaoning, China
| | - Ming Dong
- School of Stomatology, Dalian Medical University, Dalian, Liaoning, China
| | - Lina Wang
- School of Stomatology, Dalian Medical University, Dalian, Liaoning, China
| | - Qian Yang
- School of Stomatology, Dalian Medical University, Dalian, Liaoning, China
| | - Long Wang
- School of Stomatology, Dalian Medical University, Dalian, Liaoning, China
| | - Wenqing Han
- School of Stomatology, Dalian Medical University, Dalian, Liaoning, China
| | - Juhong Dong
- School of Stomatology, Dalian Medical University, Dalian, Liaoning, China
| | - Tingjiao Liu
- Department of Basic Science of Stomatology, Shanghai Stomatological Hospital, Fudan University, Shanghai, China,Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China,Tingjiao Liu Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai200003, China; Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai200003, China
| | - Ying Kong
- Department Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning, China,Ying Kong Department Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian116044, Liaoning, China;
| | - Weidong Niu
- School of Stomatology, Dalian Medical University, Dalian, Liaoning, China,CONTACT Weidong Niu School of Stomatology, Dalian Medical University, Dalian116044, Liaoning, China;
| |
Collapse
|
28
|
Wu P, Wu W, Zhang S, Han J, Liu C, Yu H, Chen X, Chen X. Therapeutic potential and pharmacological significance of extracellular vesicles derived from traditional medicinal plants. Front Pharmacol 2023; 14:1272241. [PMID: 38108066 PMCID: PMC10725203 DOI: 10.3389/fphar.2023.1272241] [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: 08/03/2023] [Accepted: 11/20/2023] [Indexed: 12/19/2023] Open
Abstract
Medicinal plants are the primary sources for the discovery of novel medicines and the basis of ethnopharmacological research. While existing studies mainly focus on the chemical compounds, there is little research about the functions of other contents in medicinal plants. Extracellular vesicles (EVs) are functionally active, nanoscale, membrane-bound vesicles secreted by almost all eukaryotic cells. Intriguingly, plant-derived extracellular vesicles (PDEVs) also have been implicated to play an important role in therapeutic application. PDEVs were reported to have physical and chemical properties similar to mammalian EVs, which are rich in lipids, proteins, nucleic acids, and pharmacologically active compounds. Besides these properties, PDEVs also exhibit unique advantages, especially intrinsic bioactivity, high stability, and easy absorption. PDEVs were found to be transferred into recipient cells and significantly affect their biological process involved in many diseases, such as inflammation and tumors. PDEVs also could offer unique morphological and compositional characteristics as natural nanocarriers by innately shuttling bioactive lipids, RNA, proteins, and other pharmacologically active substances. In addition, PDEVs could effectively encapsulate hydrophobic and hydrophilic chemicals, remain stable, and cross stringent biological barriers. Thus, this study focuses on the pharmacological action and mechanisms of PDEVs in therapeutic applications. We also systemically deal with facets of PDEVs, ranging from their isolation to composition, biological functions, and biotherapeutic roles. Efforts are also made to elucidate recent advances in re-engineering PDEVs applied as stable, effective, and non-immunogenic therapeutic applications to meet the ever-stringent demands. Considering its unique advantages, these studies not only provide relevant scientific evidence on therapeutic applications but could also replenish and inherit precious cultural heritage.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xiping Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaofeng Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
29
|
Gong L, Zhou H, Zhang Y, Wang C, Fu K, Ma C, Li Y. Preparation of Phillygenin-Hyaluronic acid composite milk-derived exosomes and its anti-hepatic fibrosis effect. Mater Today Bio 2023; 23:100804. [PMID: 37753374 PMCID: PMC10518489 DOI: 10.1016/j.mtbio.2023.100804] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/23/2023] [Accepted: 09/15/2023] [Indexed: 09/28/2023] Open
Abstract
Liver fibrosis remains a serious problem affecting the health of millions of people worldwide. Hepatic stellate cells (HSCs) are the main effector cells in liver fibrosis and their activation could lead to extracellular matrix deposition, which may aggravate the development of liver fibrosis and inflammation. Previous studies have reported the potential of Phillygenin (PHI) as a hepatoprotective agent to inhibit HSCs activation and fibrosis development. However, the poor water solubility of PHI hinders its clinical application as a potential anti-liver fibrosis therapy. Milk-derived exosomes (mEXO) serve as scalable nanocarriers for delivering chemotherapeutic agents due to their excellent biocompatibility. Here, we developed a PHI-Hyaluronic acid (HA) composite mEXO (PHI-HA-mEXO) drug delivery system, in which DSPE-PEG2000-HA was conjugated to the surface of mEXO to prepare HA-mEXO, and PHI was encapsulated into HA-mEXO to form PHI-HA-mEXO. As a specific receptor for HA, CD44 is frequently over-expressed during liver fibrosis and highly expressed on the surface of activated HSCs (aHSCs). PHI-HA-mEXO can bind to CD44 and enter aHSCs through endocytosis and release PHI. PHI-HA-mEXO drug delivery system can significantly induce aHSCs death without affecting quiescent HSCs (qHSCs) and hepatocytes. Furthermore, we carried out in vitro and in vivo experiments and found that PHI-HA-mEXO could alleviate liver fibrosis through aHSCs-targeted mechanism. In conclusion, the favorable biosafety and superior anti-hepatic fibrosis effects suggest a promising potential of PHI-HA-mEXO in the treatment of hepatic fibrosis. However, detailed pharmokinetics and dose-responsive experiments of PHI-HA-mEXO and the mechanism of mEXO loading drugs are still required before PHI-HA-mEXO can be applied clinically.
Collapse
Affiliation(s)
| | | | - Yafang Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of StandardizatAion for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Cheng Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of StandardizatAion for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ke Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of StandardizatAion for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Cheng Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of StandardizatAion for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yunxia Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of StandardizatAion for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| |
Collapse
|
30
|
Hu J, Zhu J, Chai J, Zhao Y, Luan J, Wang Y. Application of exosomes as nanocarriers in cancer therapy. J Mater Chem B 2023; 11:10595-10612. [PMID: 37927220 DOI: 10.1039/d3tb01991h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
Cancer remains the most common lethal disease in the world. Although the treatment choices for cancer are still limited, significant progress has been made over the past few years. By improving targeted drug therapy, drug delivery systems promoted the therapeutic effects of anti-cancer medications. Exosome is a kind of natural nanoscale delivery system with natural substance transport properties, good biocompatibility, and high tumor targeting, which shows great potential in drug carriers, thereby providing novel strategies for cancer therapy. In this review, we present the formation, distribution, and characteristics of exosomes. Besides, extraction and isolation techniques are discussed. We focus on the recent progress and application of exosomes in cancer therapy in four aspects: exosome-mediated gene therapy, chemotherapy, photothermal therapy, and combination therapy. The current challenges and future developments of exosome-mediated cancer therapy are also discussed. Finally, the latest advances in the application of exosomes as drug delivery carriers in cancer therapy are summarized, which provide practical value and guidance for the development of cancer therapy.
Collapse
Affiliation(s)
- Jiawei Hu
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
| | - Junfei Zhu
- China-Japan Friendship Hospital, No. 2 Sakura East Street, Chaoyang District, Beijing, China
| | - Jingjing Chai
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
| | - Yudie Zhao
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
| | - Jiajie Luan
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
| | - Yan Wang
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
| |
Collapse
|
31
|
Lucchetti D, Colella F, Artemi G, Haque S, Sgambato A, Pellicano R, Fagoonee S. Smart nano-sized extracellular vesicles for cancer therapy: Potential theranostic applications in gastrointestinal tumors. Crit Rev Oncol Hematol 2023; 191:104121. [PMID: 37690633 DOI: 10.1016/j.critrevonc.2023.104121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 01/27/2023] [Accepted: 09/04/2023] [Indexed: 09/12/2023] Open
Abstract
Extracellular vesicles (EVs) have gained tremendous interest in the search for next-generation therapeutics for the treatment of a range of pathologies, including cancer, especially due to their small size, biomolecular cargo, ability to mediate intercellular communication, high physicochemical stability, low immunogenicity and biocompatibility. The theranostic potential of EVs have been enhanced by adopting several strategies such as genetic or metabolic engineering, parental cell modification or direct functionalization to incorporate therapeutic compounds into these nanoplatforms. The smart nano-sized EVs indeed offer huge opportunities in the field of cancer, and current research is set at overcoming the existing pitfalls. Smart EVs are already being applied in the clinics despite the challenges faced. We provide, herein, an update on the technologies employed for EV functionalization in order to achieve optimal tumor cell targeting and EV tracking in vivo with bio-imaging modalities, as well as the preclinical and clinical studies making use of these modified EVs, in the context of gastrointestinal tumors.
Collapse
Affiliation(s)
- Donatella Lucchetti
- Fondazione Policlinico Universitario 'Agostino Gemelli' IRCCS, Rome, Italy; Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Filomena Colella
- Fondazione Policlinico Universitario 'Agostino Gemelli' IRCCS, Rome, Italy
| | - Giulia Artemi
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan 45142, Saudi Arabia; Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut 1102 2801, Lebanon; Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman 13306, United Arab Emirates
| | - Alessandro Sgambato
- Fondazione Policlinico Universitario 'Agostino Gemelli' IRCCS, Rome, Italy; Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy.
| | - Rinaldo Pellicano
- Gastroenterology Unit, Città della salute e della Scienza Hospital, Turin, Italy
| | - Sharmila Fagoonee
- Institute of Biostructure and Bioimaging (CNR), Molecular Biotechnology Center, Turin, Italy
| |
Collapse
|
32
|
Zhang Y, Lin Y, He J, Song S, Luo Y, Lu Y, Chen S, Wang Q, Li Y, Ren F, Guo H. Milk-derived small extracellular vesicles: a new perspective on dairy nutrition. Crit Rev Food Sci Nutr 2023:1-22. [PMID: 37819268 DOI: 10.1080/10408398.2023.2263573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
Milk contains bioactive compounds that have multiple essential benefits. Milk-derived small extracellular vesicles (M-sEVs) have emerged as novel bioactive milk components with various beneficial biological functions and broad applications. The M-sEVs from different mammalian sources have similar composition and bioactive functions. The digestive stability and biocompatibility of the M-sEVs provide a good foundation for their physiological functions. Evidence suggests that M-sEVs promote intestinal, immune, bone, neural, liver, and heart health and show therapeutic effects against cancer, indicating their potential for use in functional foods. In addition, M-sEVs can be developed as natural delivery carriers owing to their superior structural characteristics. Further studies are needed to elucidate the relationship between the specific components and functions of M-sEVs, standardize their extraction processes, and refine relevant clinical trials to advance the future applications of M-sEVs. This review summarizes the structure and composition of M-sEVs isolated from different milk sources and discusses several common extraction methods. Since the introduction of M-sEVs for digestion and absorption, studies have been conducted on their biological functions. Furthermore, we outline the theoretical industrial production route, potential application scenarios of M-sEVs, and the future perspectives of M-sEV research.
Collapse
Affiliation(s)
- Yuning Zhang
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
| | - Yingying Lin
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, PR China
- National Center of Technology Innovation for Dairy, Hohhot, PR China
| | - Jian He
- National Center of Technology Innovation for Dairy, Hohhot, PR China
| | - Sijia Song
- Food Laboratory of Zhongyuan, Luohe, PR China
| | - Yujia Luo
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
| | - Yao Lu
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
| | | | - Qingyu Wang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, PR China
| | - Yixuan Li
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, PR China
| | - Fazheng Ren
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, PR China
| | - Huiyuan Guo
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, PR China
| |
Collapse
|
33
|
Lan Q, Cao J, Bi X, Xiao X, Li D, Ai Y. Curcumin-primed periodontal ligament stem cells-derived extracellular vesicles improve osteogenic ability through the Wnt/β-catenin pathway. Front Cell Dev Biol 2023; 11:1225449. [PMID: 37842095 PMCID: PMC10568008 DOI: 10.3389/fcell.2023.1225449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 09/07/2023] [Indexed: 10/17/2023] Open
Abstract
Introduction: Curcumin has broad application prospects in the prevention and treatment of periodontal diseases. Periodontal ligament stem cell-derived extracellular vesicles (PDLSC-EV) can effectively promote periodontal tissue regeneration and possess good drug delivery capability. Superior pharmacological effects can be exerted using PDLSC-EV as a curcumin carrier. Methods: In the present study, we constructed curcumin-primed PDLSCs-derived extracellular vesicles (Cur-PDLSC-EV) from cell culture supernatants of curcumin-pretreated PDLSCs by ultracentrifugation and investigated their effects on the proliferation, migration, and osteogenic ability of PDLSCs and the corresponding downstream molecular pathways. Results: Both Cur-PDLSC-EV and PDLSC-EV promoted osteoblast proliferation and migration. Compared with PDLSC-EV, Cur-PDLSC-EV possessed a more potent pro-osteogenic ability. Moreover, the improved osteogenesis of Cur-PDLSC-EV was related to the activation of the Wnt/β-catenin signaling pathway. Conclusion: This study suggests that Cur-PDLSC-EV can promote osteogenic differentiation by activating Wnt/β-catenin, providing reference bases for the treatment of periodontal diseases.
Collapse
Affiliation(s)
- Qian Lan
- Foshan Stomatology Hospital, School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Jiadong Cao
- Foshan Stomatology Hospital, School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Xueting Bi
- Foshan Stomatology Hospital, School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Xin Xiao
- Foshan Stomatology Hospital, School of Medicine, Foshan University, Foshan, Guangdong, China
| | | | - Yilong Ai
- Foshan Stomatology Hospital, School of Medicine, Foshan University, Foshan, Guangdong, China
| |
Collapse
|
34
|
Joo HS, Suh JH, So CM, Jeon HJ, Yoon SH, Lee JM. Emerging Roles of Using Small Extracellular Vesicles as an Anti-Cancer Drug. Int J Mol Sci 2023; 24:14063. [PMID: 37762393 PMCID: PMC10531913 DOI: 10.3390/ijms241814063] [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: 08/19/2023] [Revised: 09/07/2023] [Accepted: 09/09/2023] [Indexed: 09/29/2023] Open
Abstract
Small extracellular vesicles (sEVs) are emerging as a novel therapeutic strategy for cancer therapy. Tumor-cell-derived sEVs contain biomolecules that can be utilized for cancer diagnosis. sEVs can directly exert tumor-killing effects or modulate the tumor microenvironment, leading to anti-cancer effects. In this review, the application of sEVs as a diagnostic tool, drug delivery system, and active pharmaceutical ingredient for cancer therapy will be highlighted. The therapeutic efficacies of sEVs will be compared to conventional immune checkpoint inhibitors. Additionally, this review will provide strategies for sEV engineering to enhance the therapeutic efficacies of sEVs. As a bench-to-bedside application, we will discuss approaches to encourage good-manufacturing-practice-compliant industrial-scale manufacturing and purification of sEVs.
Collapse
Affiliation(s)
| | | | | | | | | | - Jung Min Lee
- School of Life Science, Handong Global University, 558 Handong-ro, Buk-gu, Pohang 37554, Republic of Korea
| |
Collapse
|
35
|
Liu R, Liu S, Wu S, Xia M, Liu W, Wang L, Dong M, Niu W. Milk-Derived Small Extracellular Vesicles Promote Osteogenic Differentiation and Inhibit Inflammation via microRNA-21. Int J Mol Sci 2023; 24:13873. [PMID: 37762176 PMCID: PMC10531249 DOI: 10.3390/ijms241813873] [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: 08/01/2023] [Revised: 09/02/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Chronic apical periodontitis (CAP) is a disease with characteristics of inflammation and bone loss. In this study, our objective was to examine the function of small extracellular vesicles (sEVs) obtained from milk in encouraging osteogenic differentiation and inhibiting inflammation by miR-21 in CAP. The expression of miR-21 was detected using qRT-PCR in human CAP samples. The impact of miR-21 on the process of osteogenic differentiation was investigated using CCK-8, qRT-PCR, immunofluorescence staining, and Western blot analysis. The evaluation of RAW 264.7 cell polarization and the assessment of inflammatory factor expression were conducted through qRT-PCR. The influence of sEVs on MC3T3-E1 cells and RAW 264.7 cells was examined, with a particular emphasis on the involvement of miR-21. In human CAP samples, a decrease in miR-21 expression was observed. MiR-21 increased the expression of osteogenesis-related genes and M2 polarization genes while decreasing the expression of M1 polarization genes and inflammatory cytokines. Treatment with milk-derived sEVs also promoted osteogenesis and M2 polarization while inhibiting M1 polarization and inflammation. Conversely, the addition of miR-21 inhibitors resulted in opposite effects. Our results indicated that sEVs derived from milk had a positive effect on bone formation and activation of anti-inflammatory (M2) macrophages and simultaneously reduced inflammation by regulating miR-21 in CAP.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ming Dong
- School of Stomatology, Dalian Medical University, Dalian 116044, China
| | - Weidong Niu
- School of Stomatology, Dalian Medical University, Dalian 116044, China
| |
Collapse
|
36
|
Yan R, Chen H, Selaru FM. Extracellular Vesicles in Hepatocellular Carcinoma: Progress and Challenges in the Translation from the Laboratory to Clinic. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1599. [PMID: 37763719 PMCID: PMC10534795 DOI: 10.3390/medicina59091599] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 08/27/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023]
Abstract
Extracellular vesicles (EVs) play critical roles in intercellular communication by transporting bioactive cargo to recipient cells. EVs have been implicated in a range of physiological and pathological processes, including tumor progression, metastasis, immune modulation, and drug resistance. The objective of this review is to present a thorough overview of recent studies focusing on EVs in hepatocellular carcinoma (HCC), with an emphasis on their potential utility as diagnostic biomarkers as well as therapeutic agents. Initially, we explore the utility of EVs as diagnostic biomarkers for HCC, followed by a discussion of their potential as carriers of therapeutic payloads. Additionally, we delve into the emerging field of therapeutic EVs for modulating tumor immune responses. Through this review, our ultimate aim is to provide a comprehensive understanding of the opportunities and challenges in the clinical translation of EV research in the domain of HCC.
Collapse
Affiliation(s)
- Rong Yan
- Department of Surgical Oncology, the First Affiliated Hospital, Xi’an Jiaotong University College of Medicine, Xi’an 710061, China
| | - Haiming Chen
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA;
| | - Florin M. Selaru
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA;
- Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins University, Baltimore, MD 21224, USA
- The Institute for Nanobiotechnology, The Johns Hopkins University, Baltimore, MD 21231, USA
| |
Collapse
|
37
|
Mu N, Li J, Zeng L, You J, Li R, Qin A, Liu X, Yan F, Zhou Z. Plant-Derived Exosome-Like Nanovesicles: Current Progress and Prospects. Int J Nanomedicine 2023; 18:4987-5009. [PMID: 37693885 PMCID: PMC10492547 DOI: 10.2147/ijn.s420748] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/28/2023] [Indexed: 09/12/2023] Open
Abstract
Exosomes are small extracellular vesicles, ranging in size from 30-150nm, which can be derived from various types of cells. In recent years, mammalian-derived exosomes have been extensively studied and found to play a crucial role in regulating intercellular communication, thereby influencing the development and progression of numerous diseases. Traditional Chinese medicine has employed plant-based remedies for thousands of years, and an increasing body of evidence suggests that plant-derived exosome-like nanovesicles (PELNs) share similarities with mammalian-derived exosomes in terms of their structure and function. In this review, we provide an overview of recent advances in the study of PELNs and their potential implications for human health. Specifically, we summarize the roles of PELNs in respiratory, digestive, circulatory, and other diseases. Furthermore, we have extensively investigated the potential shortcomings and challenges in current research regarding the mechanism of action, safety, administration routes, isolation and extraction methods, characterization and identification techniques, as well as drug-loading capabilities. Based on these considerations, we propose recommendations for future research directions. Overall, our review highlights the potential of PELNs as a promising area of research, with broad implications for the treatment of human diseases. We anticipate continued interest in this area and hope that our summary of recent findings will stimulate further exploration into the implications of PELNs for human health.
Collapse
Affiliation(s)
- Nai Mu
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan Province, People’s Republic of China
- Geriatric Diseases Institute of Chengdu, Department of Orthopedics, Chengdu Fifth People’s Hospital, Chengdu, Sichuan Province, People’s Republic of China
| | - Jie Li
- Center for Medicine Research and Translation, Chengdu Fifth People’s Hospital, Chengdu, Sichuan Province, People’s Republic of China
| | - Li Zeng
- Department of Pharmacy, Chengdu Fifth People’s Hospital, Chengdu, Sichuan Province, People’s Republic of China
| | - Juan You
- Department of Pharmacy, Chengdu Fifth People’s Hospital, Chengdu, Sichuan Province, People’s Republic of China
| | - Rong Li
- Department of Pharmacy, Chengdu Fifth People’s Hospital, Chengdu, Sichuan Province, People’s Republic of China
| | - Anquan Qin
- Department of Pharmacy, Chengdu Fifth People’s Hospital, Chengdu, Sichuan Province, People’s Republic of China
| | - Xueping Liu
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan Province, People’s Republic of China
| | - Fang Yan
- Center for Medicine Research and Translation, Chengdu Fifth People’s Hospital, Chengdu, Sichuan Province, People’s Republic of China
- Geriatric Diseases Institute of Chengdu, Department of Geriatrics, Chengdu Fifth People’s Hospital, Chengdu, Sichuan Province, People’s Republic of China
| | - Zheng Zhou
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan Province, People’s Republic of China
- Geriatric Diseases Institute of Chengdu, Department of Orthopedics, Chengdu Fifth People’s Hospital, Chengdu, Sichuan Province, People’s Republic of China
| |
Collapse
|
38
|
Nguyen TNG, Pham CV, Chowdhury R, Patel S, Jaysawal SK, Hou Y, Xu H, Jia L, Duan A, Tran PHL, Duan W. Development of Blueberry-Derived Extracellular Nanovesicles for Immunomodulatory Therapy. Pharmaceutics 2023; 15:2115. [PMID: 37631329 PMCID: PMC10458573 DOI: 10.3390/pharmaceutics15082115] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
Over the past decade, there has been a significant expansion in the development of plant-derived extracellular nanovesicles (EVs) as an effective drug delivery system for precision therapy. However, the lack of effective methods for the isolation and characterization of plant EVs hampers progress in the field. To solve a challenge related to systemic separation and characterization in the plant-derived EV field, herein, we report the development of a simple 3D inner filter-based method that allows the extraction of apoplastic fluid (AF) from blueberry, facilitating EV isolation as well as effective downstream applications. Class I chitinase (PR-3) was found in blueberry-derived EVs (BENVs). As Class I chitinase is expressed in a wide range of plants, it could serve as a universal marker for plant-derived EVs. Significantly, the BENVs exhibit not only higher drug loading capacity than that reported for other EVs but also possess the ability to modulate the release of the proinflammatory cytokine IL-8 and total glutathione in response to oxidative stress. Therefore, the BENV is a promising edible multifunctional nano-bio-platform for future immunomodulatory therapies.
Collapse
Affiliation(s)
- Tuong Ngoc-Gia Nguyen
- School of Medicine, Faculty of Health, Deakin University, Geelong Waurn Ponds Campus, Geelong, VIC 3216, Australia; (T.N.-G.N.); (C.V.P.); (R.C.); (S.P.); (S.K.J.)
| | - Cuong Viet Pham
- School of Medicine, Faculty of Health, Deakin University, Geelong Waurn Ponds Campus, Geelong, VIC 3216, Australia; (T.N.-G.N.); (C.V.P.); (R.C.); (S.P.); (S.K.J.)
| | - Rocky Chowdhury
- School of Medicine, Faculty of Health, Deakin University, Geelong Waurn Ponds Campus, Geelong, VIC 3216, Australia; (T.N.-G.N.); (C.V.P.); (R.C.); (S.P.); (S.K.J.)
| | - Shweta Patel
- School of Medicine, Faculty of Health, Deakin University, Geelong Waurn Ponds Campus, Geelong, VIC 3216, Australia; (T.N.-G.N.); (C.V.P.); (R.C.); (S.P.); (S.K.J.)
| | - Satendra Kumar Jaysawal
- School of Medicine, Faculty of Health, Deakin University, Geelong Waurn Ponds Campus, Geelong, VIC 3216, Australia; (T.N.-G.N.); (C.V.P.); (R.C.); (S.P.); (S.K.J.)
| | - Yingchun Hou
- Laboratory of Tumor Molecular and Cellular Biology, College of Life Sciences, Shaanxi Normal University, 620 West Chang’an Avenue, Xi’an 710119, China;
| | - Huo Xu
- College of Materials and Chemical Engineering, Minjiang University, Fuzhou 350108, China; (H.X.); (L.J.)
| | - Lee Jia
- College of Materials and Chemical Engineering, Minjiang University, Fuzhou 350108, China; (H.X.); (L.J.)
| | - Andrew Duan
- School of Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC 3800, Australia;
| | - Phuong Ha-Lien Tran
- School of Medicine, Faculty of Health, Deakin University, Geelong Waurn Ponds Campus, Geelong, VIC 3216, Australia; (T.N.-G.N.); (C.V.P.); (R.C.); (S.P.); (S.K.J.)
| | - Wei Duan
- School of Medicine, Faculty of Health, Deakin University, Geelong Waurn Ponds Campus, Geelong, VIC 3216, Australia; (T.N.-G.N.); (C.V.P.); (R.C.); (S.P.); (S.K.J.)
| |
Collapse
|
39
|
Lin SW, Tsai JC, Shyong YJ. Drug delivery of extracellular vesicles: Preparation, delivery strategies and applications. Int J Pharm 2023; 642:123185. [PMID: 37391106 DOI: 10.1016/j.ijpharm.2023.123185] [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: 03/01/2023] [Revised: 06/24/2023] [Accepted: 06/26/2023] [Indexed: 07/02/2023]
Abstract
Extracellular vesicles (EV) are cell-originated vesicles exhibited with characteristics similar to the parent cells. Several studies have suggested the therapeutic potential of EV since they played as an intercellular communicator and modulate disease microenvironment, and thus EV has been widely studied in cancer management and tissue regeneration. However, merely application of EV revealed limited therapeutic outcome in different disease scenario and co-administration of drugs may be necessary to exert proper therapeutic effect. The method of drug loading into EV and efficient delivery of the formulation is therefore important. In this review, the advantages of using EV as drug delivery system compared to traditional synthetic nanoparticles will be emphasized, followed by the method of preparing EV and drug loading. The pharmacokinetic characteristics of EV was discussed, together with the review of reported delivery strategies and related application of EV in different disease management.
Collapse
Affiliation(s)
- Shang-Wen Lin
- School of Pharmacy, College of Medicine, National Cheng Kung University, No.1, University Road, Tainan City 701, Taiwan
| | - Jui-Chen Tsai
- School of Pharmacy, College of Medicine, National Cheng Kung University, No.1, University Road, Tainan City 701, Taiwan
| | - Yan-Jye Shyong
- School of Pharmacy, College of Medicine, National Cheng Kung University, No.1, University Road, Tainan City 701, Taiwan.
| |
Collapse
|
40
|
Liu D, Wang J, You W, Ma F, Sun Q, She J, He W, Yang G. A d-peptide-based oral nanotherapeutic modulates the PD-1/PD-L1 interaction for tumor immunotherapy. Front Immunol 2023; 14:1228581. [PMID: 37529049 PMCID: PMC10388715 DOI: 10.3389/fimmu.2023.1228581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 06/23/2023] [Indexed: 08/03/2023] Open
Abstract
Background PD-1/PD-L1 immune checkpoint inhibitors are currently the most commonly utilized agents in clinical practice, which elicit an immunostimulatory response to combat malignancies. However, all these inhibitors are currently administered via injection using antibody-based therapies, while there is a growing need for oral alternatives. Methods This study has developed and synthesized exosome-wrapped gold-peptide nanocomplexes with low immunogenicity, which can target PD-L1 and activate antitumor immunity in vivo through oral absorption. The SuperPDL1exo was characterized by transmission electron microscopy (TEM), dynamic light scattering (DLS), Fourier transform infrared (FTIR), X-ray photoelectron spectroscopy (XPS), and gel silver staining. The transmembrane ability of SuperPDL1exo was evaluated by flow cytometry and immunofluorescence. Cell viability was determined using the Cell Counting Kit-8 (CCK-8) assay. ELISA experiments were conducted to detect serum and tissue inflammatory factors, as well as serum biochemical indicators. Tissue sections were stained with H&E for the evaluation of the safety of SuperPDL1exo. An MC38 colon cancer model was established in immunocompetent C56BL/6 mice to evaluate the effects of SuperPDL1exo on tumor growth in vivo. Immunohistochemistry (IHC) staining was performed to detect cytotoxicity factors such as perforin and granzymes. Results First, SuperPDL1 was successfully synthesized, and milk exosome membranes were encapsulated through ultrasound, repeated freeze-thaw cycles, and extrusion, resulting in the synthesis of SuperPDL1exo. Multiple characterization results confirmed the successful synthesis of SuperPDL1exo nanoparticles. Furthermore, our data demonstrated that SuperPDL1exo exhibited excellent colloidal stability and superior cell transmembrane ability. In vitro and in vivo experiments revealed that SuperPDL1exo did not cause damage to multiple systemic organs, demonstrating its good biocompatibility. Finally, in the MC38 colon cancer mouse model, it was discovered that SuperPDL1exo could inhibit the progression of colon cancer, and this tumor-suppressive effect was mediated through the activation of tumor-specific cytotoxic T lymphocyte (CTL)-related immune responses. Conclusion This study has successfully designed and synthesized an oral nanotherapeutic, SuperPDL1exo, which demonstrates small particle size, excellent colloidal stability, transmembrane ability in tumor cells, and biocompatibility. In vivo experiments have shown that it effectively activates T-cell immunity and exerts antitumor effects.
Collapse
Affiliation(s)
- Dan Liu
- Department of General Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xian, China
- Department of Medical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Department of Talent Highland, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jingmei Wang
- Institute for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Weiming You
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Fang Ma
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Qi Sun
- Department of General Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xian, China
| | - Junjun She
- Department of General Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xian, China
| | - Wangxiao He
- Department of Medical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Department of Talent Highland, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Institute for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Guang Yang
- Department of Oncology, Kunshan Hospital of Chinese Medicine, Affiliated Hospital of Yangzhou University, Yangzhou, China
| |
Collapse
|
41
|
Franzoni G, Mecocci S, De Ciucis CG, Mura L, Dell’Anno F, Zinellu S, Fruscione F, De Paolis L, Carta T, Anfossi AG, Dei Guidici S, Chiaradia E, Pascucci L, Oggiano A, Cappelli K, Razzuoli E. Goat milk extracellular vesicles: immuno-modulation effects on porcine monocyte-derived macrophages in vitro. Front Immunol 2023; 14:1209898. [PMID: 37469517 PMCID: PMC10352104 DOI: 10.3389/fimmu.2023.1209898] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 05/26/2023] [Indexed: 07/21/2023] Open
Abstract
Introduction Extracellular vesicles (EVs) are nanometric-membrane-bound sub-cellular structures, which can be recovered from milk. Milk EVs have drawn increasing interest due to their potential biomedical applications, therefore it is important to investigate their impact on key immune cells, such as macrophages. Methods In this work, the immunomodulatory effects of goat milk EVs on untreated (moMФ) and classically activated (moM1) porcine monocyte-derived macrophages were investigated using flow cytometry, ELISA, and gene expression assays. Results These particles were efficiently internalized by macrophages and high doses (60 mg protein weight) triggered the upregulation of MHC I and MHC II DR on moMФ, but not on moM1. In moMФ, exposure to low doses (0.6 mg) of mEVs enhanced the gene expression of IL10, EBI3, and IFNB, whereas high doses up-regulated several pro-inflammatory cytokines. These nanosized structures slightly modulated cytokine gene expression on moM1. Accordingly, the cytokine (protein) contents in culture supernatants of moMФ were mildly affected by exposure to low doses of mEVs, whereas high doses promoted the increased release of TNF, IL-8, IL-1a, IL-1b, IL-1Ra, IL-6, IL-10, and IL-12. The cytokines content in moM1 supernatants was not critically affected. Discussion Overall, our data support a clinical application of these molecules: they polarized macrophages toward an M1-like phenotype, but this activation seemed to be controlled, to prevent potentially pathological over-reaction to stressors.
Collapse
Affiliation(s)
- Giulia Franzoni
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, Italy
| | - Samanta Mecocci
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | - Chiara Grazia De Ciucis
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, Genova, Italy
| | - Lorena Mura
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, Italy
- Department of Biomedical Sciences, School of Medicine, University of Sassari, Sassari, Italy
| | - Filippo Dell’Anno
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, Genova, Italy
| | - Susanna Zinellu
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, Italy
| | - Floriana Fruscione
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, Genova, Italy
| | - Livia De Paolis
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, Genova, Italy
| | - Tania Carta
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, Italy
- Department of Veterinary Medicine, University of Sassari, Sassari, Italy
| | - Antonio G. Anfossi
- Department of Veterinary Medicine, University of Sassari, Sassari, Italy
| | - Silvia Dei Guidici
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, Italy
| | | | - Luisa Pascucci
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | - Annalisa Oggiano
- Department of Animal Health, Istituto Zooprofilattico Sperimentale della Sardegna, Sassari, Italy
| | - Katia Cappelli
- Department of Veterinary Medicine, University of Perugia, Perugia, Italy
| | - Elisabetta Razzuoli
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, Genova, Italy
| |
Collapse
|
42
|
Timofeeva AM, Paramonik AP, Sedykh SS, Nevinsky GA. Milk Exosomes: Next-Generation Agents for Delivery of Anticancer Drugs and Therapeutic Nucleic Acids. Int J Mol Sci 2023; 24:10194. [PMID: 37373342 DOI: 10.3390/ijms241210194] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/05/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Exosomes are nanovesicles 40-120 nm in diameter secreted by almost all cell types and providing humoral intercellular interactions. Given the natural origin and high biocompatibility, the potential for loading various anticancer molecules and therapeutic nucleic acids inside, and the surface modification possibility for targeted delivery, exosomes are considered to be a promising means of delivery to cell cultures and experimental animal organisms. Milk is a unique natural source of exosomes available in semi-preparative and preparative quantities. Milk exosomes are highly resistant to the harsh conditions of the gastrointestinal tract. In vitro studies have demonstrated that milk exosomes have an affinity to epithelial cells, are digested by cells by endocytosis mechanism, and can be used for oral delivery. With milk exosome membranes containing hydrophilic and hydrophobic components, exosomes can be loaded with hydrophilic and lipophilic drugs. This review covers a number of scalable protocols for isolating and purifying exosomes from human, cow, and horse milk. Additionally, it considers passive and active methods for drug loading into exosomes, as well as methods for modifying and functionalizing the surface of milk exosomes with specific molecules for more efficient and specific delivery to target cells. In addition, the review considers various approaches to visualize exosomes and determine cellular localization and bio-distribution of loaded drug molecules in tissues. In conclusion, we outline new challenges for studying milk exosomes, a new generation of targeted delivery agents.
Collapse
Affiliation(s)
- Anna M Timofeeva
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 630090 Novosibirsk, Russia
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Anastasia P Paramonik
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 630090 Novosibirsk, Russia
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Sergey S Sedykh
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 630090 Novosibirsk, Russia
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Georgy A Nevinsky
- SB RAS Institute of Chemical Biology and Fundamental Medicine, 630090 Novosibirsk, Russia
- Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| |
Collapse
|
43
|
Wang L, Wang D, Ye Z, Xu J. Engineering Extracellular Vesicles as Delivery Systems in Therapeutic Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300552. [PMID: 37080941 PMCID: PMC10265081 DOI: 10.1002/advs.202300552] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/16/2023] [Indexed: 05/03/2023]
Abstract
Extracellular vesicles (EVs) are transport vesicles secreted by living cells and released into the extracellular environment. Recent studies have shown that EVs serve as "messengers" in intercellular and inter-organismal communication, in both normal and pathological processes. EVs, as natural nanocarriers, can deliver bioactivators in therapy with their endogenous transport properties. This review article describes the engineering EVs of sources, isolation method, cargo loading, boosting approach, and adjustable targeting of EVs. Furthermore, the review summarizes the recent progress made in EV-based delivery systems applications, including cancer, cardiovascular diseases, liver, kidney, nervous system diseases, and COVID-19 and emphasizes the obstacles and challenges of EV-based therapies and possible strategies.
Collapse
Affiliation(s)
- Liwei Wang
- Department of Orthopedic Surgerythe Second Affiliated HospitalZhejiang University School of MedicineHangzhou CityZhejiang Province310009P. R. China
- Orthopedics Research Institute of Zhejiang UniversityHangzhou CityZhejiang Province310009P. R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceHangzhou CityZhejiang Province310009P. R. China
- Clinical Research Center of Motor System Disease of Zhejiang ProvinceHangzhou CityZhejiang Province310009P. R. China
| | - Di Wang
- Department of Orthopedic Surgerythe Second Affiliated HospitalZhejiang University School of MedicineHangzhou CityZhejiang Province310009P. R. China
- Orthopedics Research Institute of Zhejiang UniversityHangzhou CityZhejiang Province310009P. R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceHangzhou CityZhejiang Province310009P. R. China
- Clinical Research Center of Motor System Disease of Zhejiang ProvinceHangzhou CityZhejiang Province310009P. R. China
| | - Zhaoming Ye
- Department of Orthopedic Surgerythe Second Affiliated HospitalZhejiang University School of MedicineHangzhou CityZhejiang Province310009P. R. China
- Orthopedics Research Institute of Zhejiang UniversityHangzhou CityZhejiang Province310009P. R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceHangzhou CityZhejiang Province310009P. R. China
- Clinical Research Center of Motor System Disease of Zhejiang ProvinceHangzhou CityZhejiang Province310009P. R. China
| | - Jianbin Xu
- Department of Orthopedic Surgerythe Second Affiliated HospitalZhejiang University School of MedicineHangzhou CityZhejiang Province310009P. R. China
- Orthopedics Research Institute of Zhejiang UniversityHangzhou CityZhejiang Province310009P. R. China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang ProvinceHangzhou CityZhejiang Province310009P. R. China
- Clinical Research Center of Motor System Disease of Zhejiang ProvinceHangzhou CityZhejiang Province310009P. R. China
| |
Collapse
|
44
|
Zhong Y, Wang X, Zhao X, Shen J, Wu X, Gao P, Yang P, Chen J, An W. Multifunctional Milk-Derived Small Extracellular Vesicles and Their Biomedical Applications. Pharmaceutics 2023; 15:1418. [PMID: 37242660 PMCID: PMC10223436 DOI: 10.3390/pharmaceutics15051418] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/25/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
In recent years, small extracellular vesicles (sEVs) have been regarded as the next generation of novel delivery systems after lipid nanoparticles because of their advantages and huge prospects in drug delivery. Studies have shown that sEVs are abundant in milk and therefore can be a large and economical source of sEVs. Natural milk-derived small extracellular vesicles (msEVs) have important functions such as immune regulation, anti-bacterial infection, anti-oxidative, etc., and play a beneficial role in human health at multiple levels, including intestinal health, bone/muscle metabolism, and microbiota regulation. In addition, because they can pass the gastrointestinal barrier and have low immunogenicity, good biocompatibility, and stability, msEVs are considered a crucial oral drug delivery vehicle. Moreover, msEVs can be further engineered for targeted delivery to prolong the circulation time or enhance local drug concentrations. However, msEVs separation and purification, complex contents, and quality control hinder their application in drug delivery. This paper provides a comprehensive review of the biogenesis and characteristics, isolation and purification, composition, loading methods, and function of msEVs, based on which their applications in biomedical fields are further explored.
Collapse
Affiliation(s)
- Youxiu Zhong
- Wenlin An’s Laboratory, National Vaccine & Serum Institute (NVSI), China National Biotech Group (CNBG), Sinopharm Group, No. 38 Jing Hai Second Road, Beijing 101111, China
| | - Xudong Wang
- Wenlin An’s Laboratory, National Vaccine & Serum Institute (NVSI), China National Biotech Group (CNBG), Sinopharm Group, No. 38 Jing Hai Second Road, Beijing 101111, China
| | - Xian Zhao
- Wenlin An’s Laboratory, National Vaccine & Serum Institute (NVSI), China National Biotech Group (CNBG), Sinopharm Group, No. 38 Jing Hai Second Road, Beijing 101111, China
| | - Jiuheng Shen
- Wenlin An’s Laboratory, National Vaccine & Serum Institute (NVSI), China National Biotech Group (CNBG), Sinopharm Group, No. 38 Jing Hai Second Road, Beijing 101111, China
| | - Xue Wu
- Wenlin An’s Laboratory, National Vaccine & Serum Institute (NVSI), China National Biotech Group (CNBG), Sinopharm Group, No. 38 Jing Hai Second Road, Beijing 101111, China
| | - Peifen Gao
- Wenlin An’s Laboratory, National Vaccine & Serum Institute (NVSI), China National Biotech Group (CNBG), Sinopharm Group, No. 38 Jing Hai Second Road, Beijing 101111, China
| | - Peng Yang
- Wenlin An’s Laboratory, National Vaccine & Serum Institute (NVSI), China National Biotech Group (CNBG), Sinopharm Group, No. 38 Jing Hai Second Road, Beijing 101111, China
| | - Junge Chen
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine & Shenzhen Institute of Beihang University, Beihang University, Beijing 100083, China
| | - Wenlin An
- Wenlin An’s Laboratory, National Vaccine & Serum Institute (NVSI), China National Biotech Group (CNBG), Sinopharm Group, No. 38 Jing Hai Second Road, Beijing 101111, China
| |
Collapse
|
45
|
Liang D, Liu C, Li Y, Wu C, Chen Y, Tan M, Su W. Engineering fucoxanthin-loaded probiotics' membrane vesicles for the dietary intervention of colitis. Biomaterials 2023; 297:122107. [PMID: 37058897 DOI: 10.1016/j.biomaterials.2023.122107] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 03/17/2023] [Accepted: 03/31/2023] [Indexed: 04/16/2023]
Abstract
Extracellular vesicles (EVs) are very attractive as carriers of active components due to their good immunological and their ability to penetrate the physiological barrier that synthetic delivery carriers cannot penetrate. However, the low secretion capacity of EVs limited its widespread adoption, let alone the lower yield of EVs loaded with active components. Here, we report a large-scale engineering preparation strategy of synthetic probiotic membrane vesicles for encapsulating fucoxanthin (FX-MVs), an intervention for colitis. Compared with the EVs naturally secreted by probiotics, the engineering membrane vesicles showed a 150-fold yield and richer protein. Moreover, FX-MVs improved the gastrointestinal stability of fucoxanthin and inhibited H2O2-induced oxidative damage by scavenging free radicals effectively (p < 0.05). The in vivo results showed that FX-MVs could promote the polarization of macrophages to M2 type, prevent the injury and shortening of colon tissue (p < 0.05), and improve the colonic inflammatory response. Consistently, proinflammatory cytokines were effectively suppressed after FX-MVs treatment (p < 0.05). Unexpectedly, such engineering FX-MVs could also reshape the gut microbiota communities and improve the abundance of short-chain fatty acids in the colon. This study lays a foundation for developing dietary interventions using natural foods to treat intestinal-related diseases.
Collapse
Affiliation(s)
- Duo Liang
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian, 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, Liaoning, China; State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian, 116034, Liaoning, China
| | - Chenyue Liu
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian, 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, Liaoning, China; State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian, 116034, Liaoning, China
| | - Yu Li
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian, 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, Liaoning, China; State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian, 116034, Liaoning, China
| | - Caiyun Wu
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian, 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, Liaoning, China; State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian, 116034, Liaoning, China
| | - Yuling Chen
- School of Nursing, Johns Hopkins University, Baltimore, 21205, Maryland, United States
| | - Mingqian Tan
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian, 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, Liaoning, China; State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian, 116034, Liaoning, China
| | - Wentao Su
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian, 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, Liaoning, China; State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian, 116034, Liaoning, China.
| |
Collapse
|
46
|
Kang M, Hisey C, Tsai B, Nursalim Y, Blenkiron C, Chamley LW. Placental Extracellular Vesicles Can Be Loaded with Plasmid DNA. Mol Pharm 2023; 20:1898-1913. [PMID: 36919912 PMCID: PMC11407900 DOI: 10.1021/acs.molpharmaceut.2c00533] [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] [Indexed: 03/16/2023]
Abstract
Recently, extracellular vesicles (EVs) have garnered considerable interest as potential vehicles for drug delivery, including gene therapy. Although EVs from diverse sources have been investigated, current techniques used in the field for EV generation limit large-scale EV production. The placenta is essentially a tissue transplant and has unique properties that allow it to avoid the maternal immune system making it likely that placental EVs will not generate inflammatory responses and will avoid clearance by the immune system. We propose that placental EVs produced from explant cultures are an efficient method to produce considerable quantities of EVs that would be safe to administer, and we hypothesize that placental EVs can be loaded with large exogenous plasmids. To this end, we trialed three strategies to load plasmid DNA into placental EVs, including loading via electroporation of placental tissue prior to EV isolation and loading directly into placental EVs via electroporation or direct incubation of the EVs in plasmid solution. We report that the placenta releases vast quantities of EVs compared to placental cells in monolayer cultures. We show successful loading of plasmid DNA into both large- and small-EVs following both exogenous loading strategies with more plasmid encapsulated in large-EVs. Importantly, direct incubation did not alter EV size nor quantity. Further, we showed that the loading efficiency into EVs was dependent on the exogenous plasmid DNA dose and the DNA size. These results provide realistic estimates of plasmid loading capacity into placental EVs using current technologies and showcase the potential of placental EVs as DNA delivery vehicles.
Collapse
Affiliation(s)
- Matthew Kang
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, 1023 New Zealand
| | - Colin Hisey
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, 1023 New Zealand
- Department of biomedical Engineering, The Ohio State University, Columbus, Ohio, 43210 United States
| | - Bridget Tsai
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, 1023 New Zealand
| | - Yohanes Nursalim
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, 1023 New Zealand
| | - Cherie Blenkiron
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, 1023 New Zealand
- Auckland Cancer Society Research Center (ACSRC), University of Auckland, Auckland, 1023 New Zealand
- Molecular Medicine and Pathology, University of Auckland, Auckland, 1023 New Zealand
| | - Lawrence W Chamley
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, 1023 New Zealand
| |
Collapse
|
47
|
Raguraman R, Bhavsar D, Kim D, Ren X, Sikavitsas V, Munshi A, Ramesh R. Tumor-targeted exosomes for delivery of anticancer drugs. Cancer Lett 2023; 558:216093. [PMID: 36822543 PMCID: PMC10025995 DOI: 10.1016/j.canlet.2023.216093] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/07/2023] [Accepted: 02/10/2023] [Indexed: 02/23/2023]
Abstract
Exosomes are small phospholipid bilayer vesicles that are naturally produced by all living cells, both prokaryotes and eukaryotes. The exosomes due to their unique size, reduced immunogenicity, and their ability to mimic synthetic liposomes in carrying various anticancer drugs have been tested as drug delivery vehicles for cancer treatment. An added advantage of developing exosomes as a drug carrier is the ease of manipulating their intraluminal content and their surface modification to achieve tumor-targeted drug delivery. In the past ten-years, there has been an exponential increase in the number of exosome-related studies in cancer. Preclinical studies demonstrate exosomes-mediated delivery of chemotherapeutics, biologicals and natural products produce potent anticancer activity both, in vitro and in vivo. In contrast, the number of exosome-based clinical trials are few due to challenges in the manufacturing and scalability related to large-scale production of exosomes and their storage and stability. Herein, we discuss recent advances in exosome-based drug delivery for cancer treatment in preclinical and clinical studies and conclude with challenges to be overcome for translating a larger number of exosome-based therapies into the clinic.
Collapse
Affiliation(s)
- Rajeswari Raguraman
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; OU Health Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Dhaval Bhavsar
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; OU Health Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Dongin Kim
- Department of Pharmaceutical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; OU Health Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Xiaoyu Ren
- Department of Pharmaceutical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Vassilios Sikavitsas
- School of Chemical, Biological and Material Engineering, The University of Oklahoma, Norman, Oklahoma, 73019, USA; OU Health Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Anupama Munshi
- Department of Radiation Oncology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; OU Health Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Rajagopal Ramesh
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; OU Health Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Graduate Program in Biomedical Sciences, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
48
|
Abbas MA, Al-Saigh NN, Saqallah FG. Regulation of adipogenesis by exosomal milk miRNA. Rev Endocr Metab Disord 2023; 24:297-316. [PMID: 36692804 DOI: 10.1007/s11154-023-09788-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/12/2023] [Indexed: 01/25/2023]
Abstract
Milk is a rich source of miRNA packaged in exosomes. Evidence for the systemic uptake and tissue distribution of milk exosomes was reported in newborn and adult humans and animals. Breastfeeding in infants was associated with a reduced risk of obesity. Numerous adipogenesis-related miRNAs have been detected in human milk exosomes. It has been demonstrated that ingested exosomal milk miRNAs may alter gene expression in offspring to regulate their metabolism and growth. In humans, consumption of other species' milk, such as cows and goats, is continued through adulthood. Since miRNAs are conserved, the concern of cross-species transfer of adipogenic miRNA has been raised in recent years, and the increase in obesity worldwide was attributed partially to dairy milk consumption by humans. However, evidence is still weak. Research emphasizes the need for an adequate number of exosomal milk's miRNAs to reach the target cell for biological action to be achieved. It was reported that obese women's milk had less miRNA-148a and miRNA-30b, which may affect the fat acquisition of their babies. Some exosomal milk miRNAs, such as miRNA-29, miRNA-148, miRNA-30b and miRNA-125b, may have epigenetic effects on milk recipients. Moreover, the ability of milk exosomes to cross the gastrointestinal barrier makes them a promising oral drug delivery tool. Yet, exosomes may also be tagged with specific ligands which target certain tissues. Thus, milk exosomes can be engineered and loaded with certain miRNAs responsible for adipocyte differentiation, conversion, or browning. Modifications in the miRNA cargo of exosomes can benefit human health and be an alternative to traditional drugs.
Collapse
Affiliation(s)
- Manal A Abbas
- Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman, 19328, Jordan.
- Pharmacological and Diagnostic Research Center, Al-Ahliyya Amman University, Amman, 19328, Jordan.
| | - Noor Nadhim Al-Saigh
- Department of Basic Medical Sciences, Faculty of Medicine, Ibn Sina University for Medical Siences, Amman, 11104, Jordan
| | - Fadi G Saqallah
- Discipline of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800, Penang, Malaysia
| |
Collapse
|
49
|
Melnik BC, Stadler R, Weiskirchen R, Leitzmann C, Schmitz G. Potential Pathogenic Impact of Cow’s Milk Consumption and Bovine Milk-Derived Exosomal MicroRNAs in Diffuse Large B-Cell Lymphoma. Int J Mol Sci 2023; 24:ijms24076102. [PMID: 37047075 PMCID: PMC10094152 DOI: 10.3390/ijms24076102] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/05/2023] [Accepted: 03/16/2023] [Indexed: 03/29/2023] Open
Abstract
Epidemiological evidence supports an association between cow’s milk consumption and the risk of diffuse large B-cell lymphoma (DLBCL), the most common non-Hodgkin lymphoma worldwide. This narrative review intends to elucidate the potential impact of milk-related agents, predominantly milk-derived exosomes (MDEs) and their microRNAs (miRs) in lymphomagenesis. Upregulation of PI3K-AKT-mTORC1 signaling is a common feature of DLBCL. Increased expression of B cell lymphoma 6 (BCL6) and suppression of B lymphocyte-induced maturation protein 1 (BLIMP1)/PR domain-containing protein 1 (PRDM1) are crucial pathological deviations in DLBCL. Translational evidence indicates that during the breastfeeding period, human MDE miRs support B cell proliferation via epigenetic upregulation of BCL6 (via miR-148a-3p-mediated suppression of DNA methyltransferase 1 (DNMT1) and miR-155-5p/miR-29b-5p-mediated suppression of activation-induced cytidine deaminase (AICDA) and suppression of BLIMP1 (via MDE let-7-5p/miR-125b-5p-targeting of PRDM1). After weaning with the physiological termination of MDE miR signaling, the infant’s BCL6 expression and B cell proliferation declines, whereas BLIMP1-mediated B cell maturation for adequate own antibody production rises. Because human and bovine MDE miRs share identical nucleotide sequences, the consumption of pasteurized cow’s milk in adults with the continued transfer of bioactive bovine MDE miRs may de-differentiate B cells back to the neonatal “proliferation-dominated” B cell phenotype maintaining an increased BLC6/BLIMP1 ratio. Persistent milk-induced epigenetic dysregulation of BCL6 and BLIMP1 expression may thus represent a novel driving mechanism in B cell lymphomagenesis. Bovine MDEs and their miR cargo have to be considered potential pathogens that should be removed from the human food chain.
Collapse
|
50
|
Mobahat M, Sadroddiny E, Nooshabadi VT, Ebrahimi-Barough S, Goodarzi A, Malekshahi ZV, Ai J. Curcumin-loaded human endometrial stem cells derived exosomes as an effective carrier to suppress alpha-synuclein aggregates in 6OHDA-induced Parkinson's disease mouse model. Cell Tissue Bank 2023; 24:75-91. [PMID: 35641803 DOI: 10.1007/s10561-022-10008-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 04/22/2022] [Indexed: 11/25/2022]
Abstract
Parkinson disease (PD) is considered as one of the most worldwide neurodegenerative disorders. The major reasons associated to neurodegeneration process of PD pathogenesis are oxidative stress. Many studies reported that natural antioxidant molecules, especially, curcumin can suppress inflammatory pathways and preserve dopaminergic neurons damage in PD. Further, the poor pharmacokinetics, instability of chemical structure because of fast hydrolytic degradation at physiologic condition and especially, the presence of the blood brain barrier (BBB) has regarded as a considerable restriction factor for transfer of neurotherapeutic molecules to the brain tissue. The present research aims to the fabrication of nanoformulated curcumin loaded human endometrial stem cells derived exosomes (hEnSCs EXOs-Cur) to study on enhancing curcumin penetration to the brain across BBB and to improve anti- Parkinsonism effects of curcumin against neural death and alpha-synuclein aggregation. hEnSCs EXOs-Cur characterization results demonstrated the accurate size and morphology of formulated curcumin loaded exosomes with a proper stability and sustained release profile. In vivo studies including behavioral, Immunohistochemical and molecular evaluations displayed that novel formulation of hEnSCs EXO-Cur is able to cross BBB, enhance motor uncoordinated movements, suppress the aggregation of αS protein and rescue neuronal cell death through elevation of BCL2 expression level as an anti-apoptotic protein and the expression level reduction of BAX and Caspase 3 as apoptotic markers.
Collapse
Affiliation(s)
- Mahsa Mobahat
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Esmaeil Sadroddiny
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Vajihe Taghdiri Nooshabadi
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Semnan University of Medical Science, Semnan, Iran
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering and Applied Cell Sciences, , School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Arash Goodarzi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Shiraz, Iran
| | - Ziba Veisi Malekshahi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, , School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|