1
|
Lin J, Huang X, Zhang J, Yang W, Sun F, Huang B, Lu W, Wang X. Amniotic fluid-derived exosomal miR-146a-5p ameliorates preeclampsia phenotypes by inhibiting HIF-1α/FLT-1 expression. Placenta 2025; 162:35-44. [PMID: 39987849 DOI: 10.1016/j.placenta.2025.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 02/25/2025]
Abstract
INTRODUCTION Preeclampsia (PE) is a pregnancy-specific complication that begins with hypertension and proteinuria and seriously threatens the health of pregnant women and fetuses. Abnormal expression of amniotic fluid-derived exosomal miR-146a-5p was observed in PE. However, the role of human amniotic fluid-derived exosomes (AF-Exos) and miR-146a-5p in PE remains unclear. METHODS We determined the miR-146a-5p expression pattern in the AF-Exos. AF-Exos, Cobalt chloride (CoCl2) and miR-146a-5p mimic were added to trophoblast cell lines HTR-8/SVneo and JEG-3, respectively. Then the proliferation and migration function of HTR-8/SVneo and JEG-3 cells were examined. The expression of miR-146a-5p, HIF-1α and FLT-1 in HTR-8/SVneo and JEG-3 cells were detected by RT-qPCR and western blotting. Finally, we determined the effect of AF-Exos in PE rat models. RESULTS MiR-146a-5p was down-regulated in AF-Exos of PE compared to normal. Co-cultured with normal AF-Exos significantly promoted proliferation and migration of HTR-8/SVneo and JEG-3 cells. CoCl2 inhibited proliferation and migration of HTR-8/SVneo and JEG-3 cells, while miR-146a-5p mimic reversed them by suppressing HIF-1α/FLT-1 expression. After treatment of AF-Exos, the blood pressure and 24-h urinary protein of PE rats were substantially decreased, the quality of fetuses and placenta exhibited improved, and HIF-1α/FLT-1 expression of placenta, sFlt-1 and sEng levels of blood, were substantial suppressed. CONCLUSION The study provided experimental evidence for the protective effects of normal AF-Exos on ameliorating preeclampsia phenotypes, and miR-146a-5p may act an important role in enhancing the proliferation and migration of trophoblast cells by targeting HIF-1α.
Collapse
Affiliation(s)
- Jin Lin
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Xiaohong Huang
- Department of Pathology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Jing Zhang
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Weiming Yang
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Fan Sun
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Bo Huang
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Wan Lu
- Jiangxi Provincial Key Laboratory of Birth Defect for Prevention and Control, Medical Genetics Center, Jiangxi Maternal and Child Health Hospital, Nanchang, 330006, China.
| | - Xiaozhong Wang
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
2
|
Xu W, Jieda X, Wu Y, Du F, Ma L, Luo L, Liu D, Guo L, Liu J, Dong W. Safety, Efficacy and Bio-Distribution Analysis of Exosomes Derived From Human Umbilical Cord Mesenchymal Stem Cells for Effective Treatment of Bronchopulmonary Dysplasia by Intranasal Administration in Mice Model. Int J Nanomedicine 2025; 20:2521-2553. [PMID: 40034220 PMCID: PMC11874997 DOI: 10.2147/ijn.s501843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 02/04/2025] [Indexed: 03/05/2025] Open
Abstract
Purpose Exosomes (Exos) derived from human umbilical cord mesenchymal stem cells (hUC-MSCs) hold great potential for treating bronchopulmonary dysplasia (BPD); however, safety concerns and effects of intranasal administration remain unexplored. This study aimed to explore the safety of hUC-MSCs and Exos and to investigate the efficacy and bio-distribution of repeated intranasal Exos administration in neonatal BPD models. Methods Characteristics of hUC-MSCs and Exos were analyzed. A subcutaneous tumor formation assay using a single dose of hUC-MSCs or Exos was conducted in Crl:NU-Foxn1nu mice. Vital signs, biochemical indices, pathological alterations, and 18F-FDG microPET/CT analysis were examined. Pulmonary pathology, three-dimensional reconstructions, ultrastructural structures, in vivo and ex vivo bio-distribution imaging analyses, enzyme-linked immunoassay assays, and reverse transcription-quantitative polymerase chain reaction analyses of lung tissues were all documented following intranasal Exos administration. Results Characteristics of hUC-MSCs and Exos satisfied specifications. Crl:NU-Foxn1nu mice did not exhibit overt toxicity or carcinogenicity following a single dose of hUC-MSCs or Exos after 60 days of observation. Repeated intranasal Exos administration effectively alleviated pathological injuries, restored pulmonary ventilation in three-dimensional reconstruction, and recovered endothelial cell layer integrity in ultrastructural analysis. Exos steadily accumulated in lung tissues from postnatal day 1 to 14. Exos also interrupted the epithelial-mesenchymal transition and inflammation reactions in BPD models. Conclusion As a nanoscale, non-cellular therapy, intranasal administration of Exos was an effective, noninvasive treatment for BPD. This approach was free from toxic, tumorigenic risks and repaired alveolar damage while interrupting epithelial-mesenchymal transition and inflammation in neonatal mice with BPD.
Collapse
Affiliation(s)
- Wanting Xu
- Division of Neonatology, Department of Pediatrics, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Xiaolin Jieda
- Division of Neonatology, Department of Pediatrics, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Yue Wu
- Division of Neonatology, Department of Pediatrics, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Fengling Du
- Division of Neonatology, Department of Pediatrics, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Lu Ma
- Division of Neonatology, Department of Pediatrics, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Lijuan Luo
- Division of Neonatology, Department of Pediatrics, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Dong Liu
- Division of Neonatology, Department of Pediatrics, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Ling Guo
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Jing Liu
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Wenbin Dong
- Division of Neonatology, Department of Pediatrics, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People’s Republic of China
| |
Collapse
|
3
|
Bowen CM, Ditmars F, Liu N, Abril JM, Ajasin D, Russell WK, Stevenson H, Eugenin EA, Fair JH, Fagg WS. Amniotic Fluid Reduces Liver Fibrosis By Attenuating Hepatic Stellate Cell Activation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.20.639215. [PMID: 40027749 PMCID: PMC11870538 DOI: 10.1101/2025.02.20.639215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Regardless of the source of injury or metabolic dysfunction, fibrosis is a frequent driver of liver pathology. Excessive liver fibrosis is caused by persistent activation of hepatic stellate cells (HSCs), which is defined by myofibroblast activation (MFA) and the epithelial-mesenchymal transition (EMT). Strategies to prevent or reverse this HSC phenotype will be critical for successful treatment of liver fibrosis. We have previously shown that full-term, cell-free human amniotic fluid (cfAF) inhibits MFA and EMT in fibroblasts in vitro. We hypothesize that cfAF treatment can attenuate HSC activation and limit liver fibrosis. We tested if cfAF could prevent liver fibrosis or HSC activation in murine models of liver damage, three-dimensional hepatic spheroids, and HSC cultures. Administering cfAF prevented weight loss and the extent of fibrosis in mice with chronic liver damage without stimulating deleterious immune responses. Gene expression profiling and immunostaining indicated that cfAF administration in carbon tetrachloride-treated mice reduced EMT- and MFA-related biomarker abundance and modulated transcript levels associated with liver metabolism, immune regulatory pathways, and cell signaling. cfAF treatment lowered MFA biomarker levels in a dose-dependent manner in hepatic spheroids exposed to ethanol. Treating HSCs with cfAF in vitro strongly repressed EMT. Multi-omics analyses revealed that it also attenuates TGFβ-induced MFA and inflammation-associated processes. Thus, cfAF treatment prevents liver fibrosis by safeguarding against persistent HSC activation. These findings suggest that cfAF may be a safe and effective therapy for reducing liver fibrosis and preventing the development of cirrhosis and/or hepatocellular carcinoma.
Collapse
Affiliation(s)
- Charles M. Bowen
- Division of Transplant, Department of Surgery, University of Texas Medical Branch, Galveston, Texas, 77555, USA
- John Sealy School of Medicine, University of Texas Medical Branch, Galveston, Texas, 77555, USA
| | - Frederick Ditmars
- Division of Transplant, Department of Surgery, University of Texas Medical Branch, Galveston, Texas, 77555, USA
- John Sealy School of Medicine, University of Texas Medical Branch, Galveston, Texas, 77555, USA
| | - Naiyou Liu
- Division of Transplant, Department of Surgery, University of Texas Medical Branch, Galveston, Texas, 77555, USA
| | - Jose Marri Abril
- Division of Transplant, Department of Surgery, University of Texas Medical Branch, Galveston, Texas, 77555, USA
- John Sealy School of Medicine, University of Texas Medical Branch, Galveston, Texas, 77555, USA
| | - David Ajasin
- Department of Neurobiology, Cell Biology, and Anatomy, University of Texas Medical Branch, Galveston, Texas, 77555, USA
| | - William K. Russell
- Deparment of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, 77555, USA
| | - Heather Stevenson
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, 77555, USA
| | - Eliseo A. Eugenin
- Department of Neurobiology, Cell Biology, and Anatomy, University of Texas Medical Branch, Galveston, Texas, 77555, USA
| | - Jeffrey H. Fair
- Division of Transplant, Department of Surgery, University of Texas Medical Branch, Galveston, Texas, 77555, USA
| | - W. Samuel Fagg
- Division of Transplant, Department of Surgery, University of Texas Medical Branch, Galveston, Texas, 77555, USA
- Deparment of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, 77555, USA
- Merakris Therapeutics, RTP Frontier, Research Triangle Park, North Carolina, 27709, USA
| |
Collapse
|
4
|
Rahnama M, Heidari M, Poursalehi Z, Golchin A. Global Trends of Exosomes Application in Clinical Trials: A Scoping Review. Stem Cell Rev Rep 2024; 20:2165-2193. [PMID: 39340738 DOI: 10.1007/s12015-024-10791-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND Exosomes, nano-sized extracellular vesicles, have emerged as a promising tool for the diagnosis and treatment of various intractable diseases, including chronic wounds and cancers. As our understanding of exosomes continues to grow, their potential as a powerful therapeutic modality in medicine is also expanding. This systematic review aims to examine the progress of exosome-based clinical trials and provide a comprehensive overview of the therapeutic perspectives of exosomes. METHODS This systematic review strictly follows PRISMA guidelines and has been registered in PROSPERO, the International Prospective Register of Systematic Reviews. It encompasses articles from January 2000 to January 2023, sourced from bibliographic databases, with targeted search terms targeting exosome applications in clinical trials. During the screening process, strict inclusion and exclusion criteria were applied, including a focus on clinical trials utilizing different cell-derived exosomes for therapeutic purposes. RESULTS Among the 522 publications initially identified, only 10 studies met the stringent eligibility criteria after meticulous screening. The selection process involved systematically excluding duplicates and irrelevant articles to provide a transparent overview. CONCLUSION According to our systematic review, exosomes have promising applications in a variety of medical fields, including cell-free therapies and drug delivery systems for treating a variety of diseases, especially cancers and chronic wounds. To ensure safety, potency, and broader clinical applications, further optimization of exosome extraction, loading, targeting, and administration is necessary. While cell-based therapeutics are increasingly utilizing exosomes, this field is still in its infancy, and ongoing clinical trials will provide valuable insights into the clinical utility of exosomes.
Collapse
Affiliation(s)
- Maryam Rahnama
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
- Department of Applied Cell Sciences, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mohammad Heidari
- Department of Biostatistics and Epidemiology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Zahra Poursalehi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
- Department of Applied Cell Sciences, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Ali Golchin
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran.
- Department of Applied Cell Sciences, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
5
|
Selvadoss A, Baby HM, Zhang H, Bajpayee AG. Harnessing exosomes for advanced osteoarthritis therapy. NANOSCALE 2024; 16:19174-19191. [PMID: 39323205 PMCID: PMC11799831 DOI: 10.1039/d4nr02792b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 09/15/2024] [Indexed: 09/27/2024]
Abstract
Exosomes are nanosized, lipid membrane vesicles secreted by cells, facilitating intercellular communication by transferring cargo from parent to recipient cells. This capability enables biological crosstalk across multiple tissues and cells. Extensive research has been conducted on their role in the pathogenesis of degenerative musculoskeletal diseases such as osteoarthritis (OA), a chronic and painful joint disease that particularly affects cartilage. Currently, no effective treatment exists for OA. Given that exosomes naturally modulate synovial joint inflammation and facilitate cartilage matrix synthesis, they are promising candidates as next generation nanocarriers for OA therapy. Recent advancements have focused on engineering exosomes through endogenous and exogenous approaches to enhance their joint retention, cartilage and chondrocyte targeting properties, and therapeutic content enrichment, further increasing their potential for OA drug delivery. Notably, charge-reversed exosomes that utilize electrostatic binding interactions with cartilage anionic aggrecan glycosaminoglycans have demonstrated the ability to penetrate the full thickness of early-stage arthritic cartilage tissue following intra-articular administration, maximizing their therapeutic potential. These exosomes offer a non-viral, naturally derived, cell-free carrier for OA drug and gene delivery applications. Efforts to standardize exosome harvest, engineering, and property characterization methods, along with scaling up production, will facilitate more efficient and rapid clinical translation. This article reviews the current state-of-the-art, explores opportunities for exosomes as OA therapeutics, and identifies potential challenges in their clinical translation.
Collapse
Affiliation(s)
- Andrew Selvadoss
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA.
| | - Helna M Baby
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Hengli Zhang
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Ambika G Bajpayee
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA.
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
6
|
Loeffler DA. Enhancing of cerebral Abeta clearance by modulation of ABC transporter expression: a review of experimental approaches. Front Aging Neurosci 2024; 16:1368200. [PMID: 38872626 PMCID: PMC11170721 DOI: 10.3389/fnagi.2024.1368200] [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: 01/10/2024] [Accepted: 05/01/2024] [Indexed: 06/15/2024] Open
Abstract
Clearance of amyloid-beta (Aβ) from the brain is impaired in both early-onset and late-onset Alzheimer's disease (AD). Mechanisms for clearing cerebral Aβ include proteolytic degradation, antibody-mediated clearance, blood brain barrier and blood cerebrospinal fluid barrier efflux, glymphatic drainage, and perivascular drainage. ATP-binding cassette (ABC) transporters are membrane efflux pumps driven by ATP hydrolysis. Their functions include maintenance of brain homeostasis by removing toxic peptides and compounds, and transport of bioactive molecules including cholesterol. Some ABC transporters contribute to lowering of cerebral Aβ. Mechanisms suggested for ABC transporter-mediated lowering of brain Aβ, in addition to exporting of Aβ across the blood brain and blood cerebrospinal fluid barriers, include apolipoprotein E lipidation, microglial activation, decreased amyloidogenic processing of amyloid precursor protein, and restricting the entrance of Aβ into the brain. The ABC transporter superfamily in humans includes 49 proteins, eight of which have been suggested to reduce cerebral Aβ levels. This review discusses experimental approaches for increasing the expression of these ABC transporters, clinical applications of these approaches, changes in the expression and/or activity of these transporters in AD and transgenic mouse models of AD, and findings in the few clinical trials which have examined the effects of these approaches in patients with AD or mild cognitive impairment. The possibility that therapeutic upregulation of ABC transporters which promote clearance of cerebral Aβ may slow the clinical progression of AD merits further consideration.
Collapse
Affiliation(s)
- David A. Loeffler
- Department of Neurology, Beaumont Research Institute, Corewell Health, Royal Oak, MI, United States
| |
Collapse
|
7
|
Ahmed SH, AlMoslemany MA, Witwer KW, Tehamy AG, El-Badri N. Stem Cell Extracellular Vesicles as Anti-SARS-CoV-2 Immunomodulatory Therapeutics: A Systematic Review of Clinical and Preclinical Studies. Stem Cell Rev Rep 2024; 20:900-930. [PMID: 38393666 PMCID: PMC11087360 DOI: 10.1007/s12015-023-10675-2] [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] [Accepted: 12/28/2023] [Indexed: 02/25/2024]
Abstract
BACKGROUND COVID-19 rapidly escalated into a worldwide pandemic with elevated infectivity even from asymptomatic patients. Complications can lead to severe pneumonia and acute respiratory distress syndrome (ARDS), which are the main contributors to death. Because of their regenerative and immunomodulatory capacities, stem cells and their derived extracellular vesicles (EVs) are perceived as promising therapies against severe pulmonary conditions, including those associated with COVID-19. Herein, we evaluate the safety and efficacy of stem cell EVs in treating COVID-19 and complicating pneumonia, acute lung injury, and ARDS. We also cover relevant preclinical studies to recapitulate the current progress in stem cell EV-based therapy. METHODS Using PubMed, Cochrane Central Register of Controlled Trials, Scopus, and Web of Science, we searched for all English-language published studies (2000-2023) that used stem cell EVs as a therapy for COVID-19, ARDS, or pneumonia. The risk of bias (ROB) was assessed for all studies. RESULTS Forty-eight studies met our inclusion criteria. Various-sized EVs derived from different types of stem cells were reported as a potentially safe and effective therapy to attenuate the cytokine storm induced by COVID-19. EVs alleviated inflammation and regenerated the alveolar epithelium by decreasing apoptosis, proinflammatory cytokines, neutrophil infiltration, and M2 macrophage polarization. They also prevented fibrin production and promoted the production of anti-inflammatory cytokines and endothelial cell junction proteins. CONCLUSION Similar to their parental cells, stem cell EVs mediate lung tissue regeneration by targeting multiple pathways and thus hold promise in promoting the recovery of COVID-19 patients and improving the survival rate of severely affected patients.
Collapse
Affiliation(s)
- Sarah Hamdy Ahmed
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, Giza, 6th of October City, 12582, Egypt
- Biotechnology/Biomolecular Chemistry Department, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Mohamed Atef AlMoslemany
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, Giza, 6th of October City, 12582, Egypt
| | - Kenneth Whitaker Witwer
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology and Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Richman Family Precision Medicine Center of Excellence in Alzheimer's Disease, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ahmed Gamal Tehamy
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, Giza, 6th of October City, 12582, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, October Gardens, Giza, 6th of October City, 12582, Egypt.
| |
Collapse
|
8
|
Fusco C, De Rosa G, Spatocco I, Vitiello E, Procaccini C, Frigè C, Pellegrini V, La Grotta R, Furlan R, Matarese G, Prattichizzo F, de Candia P. Extracellular vesicles as human therapeutics: A scoping review of the literature. J Extracell Vesicles 2024; 13:e12433. [PMID: 38738585 PMCID: PMC11089593 DOI: 10.1002/jev2.12433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 03/03/2024] [Accepted: 03/27/2024] [Indexed: 05/14/2024] Open
Abstract
Extracellular vesicles (EVs) are released by all cells and contribute to cell-to-cell communication. The capacity of EVs to target specific cells and to efficiently deliver a composite profile of functional molecules have led researchers around the world to hypothesize their potential as therapeutics. While studies of EV treatment in animal models are numerous, their actual clinical benefit in humans has more slowly started to be tested. In this scoping review, we searched PubMed and other databases up to 31 December 2023 and, starting from 13,567 records, we selected 40 pertinent published studies testing EVs as therapeutics in humans. The analysis of those 40 studies shows that they are all small pilot trials with a large heterogeneity in terms of administration route and target disease. Moreover, the absence of a placebo control in most of the studies, the predominant local application of EV formulations and the inconsistent administration dose metric still impede comparison across studies and firm conclusions about EV safety and efficacy. On the other hand, the recording of some promising outcomes strongly calls out for well-designed larger studies to test EVs as an alternative approach to treat human diseases with no or few therapeutic options.
Collapse
Affiliation(s)
- Clorinda Fusco
- Dipartimento di Medicina Molecolare e Biotecnologie MedicheUniversità degli Studi di Napoli Federico IINaplesItaly
| | - Giusy De Rosa
- Dipartimento di Medicina Molecolare e Biotecnologie MedicheUniversità degli Studi di Napoli Federico IINaplesItaly
| | - Ilaria Spatocco
- Dipartimento di Medicina Molecolare e Biotecnologie MedicheUniversità degli Studi di Napoli Federico IINaplesItaly
| | - Elisabetta Vitiello
- Dipartimento di Medicina Molecolare e Biotecnologie MedicheUniversità degli Studi di Napoli Federico IINaplesItaly
| | - Claudio Procaccini
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia SperimentaleConsiglio Nazionale delle Ricerche (IEOS‐CNR)NaplesItaly
- Unità di Neuroimmunologia, Fondazione Santa LuciaRomeItaly
| | | | | | | | - Roberto Furlan
- Clinical Neuroimmunology Unit, Institute of Experimental Neurology, Division of NeuroscienceIRCCS Ospedale San RaffaeleMilanItaly
| | - Giuseppe Matarese
- Dipartimento di Medicina Molecolare e Biotecnologie MedicheUniversità degli Studi di Napoli Federico IINaplesItaly
- Laboratorio di Immunologia, Istituto per l'Endocrinologia e l'Oncologia SperimentaleConsiglio Nazionale delle Ricerche (IEOS‐CNR)NaplesItaly
| | | | - Paola de Candia
- Dipartimento di Medicina Molecolare e Biotecnologie MedicheUniversità degli Studi di Napoli Federico IINaplesItaly
| |
Collapse
|
9
|
Atukorala I, Hannan N, Hui L. Immersed in a reservoir of potential: amniotic fluid-derived extracellular vesicles. J Transl Med 2024; 22:348. [PMID: 38609955 PMCID: PMC11010396 DOI: 10.1186/s12967-024-05154-2] [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: 11/08/2023] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
This review aims to encapsulate the current knowledge in extracellular vesicles extracted from amniotic fluid and amniotic fluid derived stem/stromal cells. Amniotic fluid (AF) bathes the developing fetus, providing nutrients and protection from biological and mechanical dangers. In addition to containing a myriad of proteins, immunoglobulins and growth factors, AF is a rich source of extracellular vesicles (EVs). These vesicles originate from cells in the fetoplacental unit. They are biological messengers carrying an active cargo enveloped within the lipid bilayer. EVs in reproduction are known to play key roles in all stages of pregnancy, starting from fertilisation through to parturition. The intriguing biology of AF-derived EVs (AF-EVs) in pregnancy and their untapped potential as biomarkers is currently gaining attention. EV studies in numerous animal and human disease models have raised expectations of their utility as therapeutics. Amniotic fluid stem cell and mesenchymal stromal cell-derived EVs (AFSC-EVs) provide an established supply of laboratory-made EVs. This cell-free mode of therapy is popular as an alternative to stem cell therapy, revealing similar, if not better therapeutic outcomes. Research has demonstrated the successful application of AF-EVs and AFSC-EVs in therapy, harnessing their anti-inflammatory, angiogenic and regenerative properties. This review provides an overview of such studies and discusses concerns in this emerging field of research.
Collapse
Affiliation(s)
- Ishara Atukorala
- Department of Obstetrics, Gynaecology & Newborn Health, Melbourne Medical School, The University of Melbourne, Mercy Hospital for Women, 163 Studley Road, Heidelberg, VIC, 3084, Australia.
- Department of Obstetrics, Gynaecology & Newborn Health, The Northern Centre for Health Education and Research, Northern Health, Epping, VIC, Australia.
| | - Natalie Hannan
- Department of Obstetrics, Gynaecology & Newborn Health, Melbourne Medical School, The University of Melbourne, Mercy Hospital for Women, 163 Studley Road, Heidelberg, VIC, 3084, Australia
- Department of Obstetrics, Gynaecology & Newborn Health, The Northern Centre for Health Education and Research, Northern Health, Epping, VIC, Australia
| | - Lisa Hui
- Department of Obstetrics, Gynaecology & Newborn Health, Melbourne Medical School, The University of Melbourne, Mercy Hospital for Women, 163 Studley Road, Heidelberg, VIC, 3084, Australia
- Department of Obstetrics, Gynaecology & Newborn Health, The Northern Centre for Health Education and Research, Northern Health, Epping, VIC, Australia
- Department of Perinatal Medicine, Mercy Hospital for Women, Mercy Health, Heidelberg, VIC, Australia
- Reproductive Epidemiology Group, Murdoch Children's Research Institute, Parkville, VIC, Australia
| |
Collapse
|
10
|
Chanda D, Del Rivero T, Ghimire R, More S, Mitrani MI, Bellio MA, Channappanavar R. Acellular Human Amniotic Fluid-Derived Extracellular Vesicles as Novel Anti-Inflammatory Therapeutics against SARS-CoV-2 Infection. Viruses 2024; 16:273. [PMID: 38400048 PMCID: PMC10892347 DOI: 10.3390/v16020273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
The ongoing COVID-19 pandemic caused by SARS-CoV-2 is associated with acute respiratory distress syndrome (ARDS) and fatal pneumonia. Excessive inflammation caused by SARS-CoV-2 is the key driver of ARDS and lethal disease. Several FDA-approved drugs that suppress virus replication are in clinical use. However, despite strong evidence for the role of virus-induced inflammation in severe COVID-19, no effective anti-inflammatory drug is available to control fatal inflammation as well as efficiently clear the virus. Therefore, there is an urgent need to identify biologically derived immunomodulators that suppress inflammation and promote antiviral immunity. In this study, we evaluated acellular human amniotic fluid (acAF) containing extracellular vesicles (hAF-EVs) as a potential non-toxic and safe biologic for immunomodulation during COVID-19. Our in vitro results showed that acAF significantly reduced inflammatory cytokine production in TLR2/4/7 and SARS-CoV-2 structural protein-stimulated mouse macrophages. Importantly, an intraperitoneal administration of acAF reduced morbidity and mortality in SARS-CoV-2-infected mice. A detailed examination of SARS-CoV-2-infected lungs revealed that the increased protection in acAF-treated mice was associated with reduced viral titers and levels of inflammatory myeloid cell infiltration. Collectively, our results identify a novel biologic that has potential to suppress excessive inflammation and enhance survival following SARS-CoV-2 infection, highlighting the translational potential of acAF against COVID-19.
Collapse
Affiliation(s)
- Debarati Chanda
- Department of Veterinary Pathobiology, Oklahoma State University, Stillwater, OK 74078, USA; (D.C.); (R.G.); (S.M.)
| | - Tania Del Rivero
- Organicell Regenerative Medicine, Davie, FL 33314, USA; (T.D.R.); (M.I.M.)
| | - Roshan Ghimire
- Department of Veterinary Pathobiology, Oklahoma State University, Stillwater, OK 74078, USA; (D.C.); (R.G.); (S.M.)
| | - Sunil More
- Department of Veterinary Pathobiology, Oklahoma State University, Stillwater, OK 74078, USA; (D.C.); (R.G.); (S.M.)
| | - Maria Ines Mitrani
- Organicell Regenerative Medicine, Davie, FL 33314, USA; (T.D.R.); (M.I.M.)
| | - Michael A. Bellio
- Organicell Regenerative Medicine, Davie, FL 33314, USA; (T.D.R.); (M.I.M.)
| | - Rudragouda Channappanavar
- Department of Veterinary Pathobiology, Oklahoma State University, Stillwater, OK 74078, USA; (D.C.); (R.G.); (S.M.)
| |
Collapse
|
11
|
Liang D, Liu C, Yang M. Mesenchymal stem cells and their derived exosomes for ALI/ARDS: A promising therapy. Heliyon 2023; 9:e20387. [PMID: 37842582 PMCID: PMC10568335 DOI: 10.1016/j.heliyon.2023.e20387] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 09/01/2023] [Accepted: 09/20/2023] [Indexed: 10/17/2023] Open
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a serious clinical syndrome with a high morbidity and mortality. Presently, therapeutic approaches for ALI/ARDS primarily revolve around symptomatic supportive care encompassing mechanical ventilation and fluid management. Regrettably, the prognosis for most ALI/ARDS patients remains bleak due to the absence of effective treatment strategies. Even survivors of ALI/ARDS may have long-term pulmonary dysfunction and cognitive impairment. The quality of life has been seriously compromised. The emergence of mesenchymal stem cells (MSCs) and their exosomes has opened up an expansive realm of potential and optimism for addressing the plight of ALI/ARDS patients, as MSCs and their derived exosomes exhibit multifaceted capabilities, including anti-inflammatory properties, facilitation of tissue repair and regeneration, and apoptosis inhibition. Therefore, future research should focus on the possible mechanisms of MSCs and their derived exosomes for the treatment of ALI/ARDS and open up new avenues for their clinical applications.
Collapse
Affiliation(s)
- Dan Liang
- Department of Endocrine, The First People's Hospital of Chongqing Liangjiang New Area, Chongqing, China
| | - Chang Liu
- School of Medicine, Nankai University, Tianjin, China
| | - Mei Yang
- Department of Endocrine, The First People's Hospital of Chongqing Liangjiang New Area, Chongqing, China
| |
Collapse
|
12
|
Mori T, Giovannelli L, Bilia AR, Margheri F. Exosomes: Potential Next-Generation Nanocarriers for the Therapy of Inflammatory Diseases. Pharmaceutics 2023; 15:2276. [PMID: 37765245 PMCID: PMC10537720 DOI: 10.3390/pharmaceutics15092276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 08/28/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
Inflammatory diseases are common pathological processes caused by various acute and chronic factors, and some of them are autoimmune diseases. Exosomes are fundamental extracellular vesicles secreted by almost all cells, which contain a series of constituents, i.e., cytoskeletal and cytosolic proteins (actin, tubulin, and histones), nucleic acids (mRNA, miRNA, and DNA), lipids (diacylglycerophosphates, cholesterol, sphingomyelin, and ceramide), and other bioactive components (cytokines, signal transduction proteins, enzymes, antigen presentation and membrane transport/fusion molecules, and adhesion molecules). This review will be a synopsis of the knowledge on the contribution of exosomes from different cell sources as possible therapeutic agents against inflammation, focusing on several inflammatory diseases, neurological diseases, rheumatoid arthritis and osteoarthritis, intestinal bowel disease, asthma, and liver and kidney injuries. Current knowledge indicates that the role of exosomes in the therapy of inflammation and in inflammatory diseases could be distinctive. The main limitations to their clinical translation are still production, isolation, and storage. Additionally, there is an urgent need to personalize the treatments in terms of the selection of exosomes; their dosages and routes of administration; and a deeper knowledge about their biodistribution, type and incidence of adverse events, and long-term effects of exosomes. In conclusion, exosomes can be a very promising next-generation therapeutic option, superior to synthetic nanocarriers and cell therapy, and can represent a new strategy of effective, safe, versatile, and selective delivery systems in the future.
Collapse
Affiliation(s)
- Tosca Mori
- Department of Chemistry “Ugo Schiff” (DICUS), University of Florence, Via Ugo Schiff 6, Sesto Fiorentino, 50019 Florence, Italy;
| | - Lisa Giovannelli
- Department of Neurosciences (Department of Neurosciences, Psychology, Drug Research and Child Health), University of Florence, 50139 Florence, Italy
| | - Anna Rita Bilia
- Department of Chemistry “Ugo Schiff” (DICUS), University of Florence, Via Ugo Schiff 6, Sesto Fiorentino, 50019 Florence, Italy;
| | - Francesca Margheri
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50121 Florence, Italy;
| |
Collapse
|
13
|
Park HS, Chugh RM, Seok J, Cetin E, Mohammed H, Siblini H, Liakath Ali F, Ghasroldasht MM, Alkelani H, Elsharoud A, Ulin M, Esfandyari S, Al-Hendy A. Comparison of the therapeutic effects between stem cells and exosomes in primary ovarian insufficiency: as promising as cells but different persistency and dosage. Stem Cell Res Ther 2023; 14:165. [PMID: 37340468 DOI: 10.1186/s13287-023-03397-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 06/05/2023] [Indexed: 06/22/2023] Open
Abstract
BACKGROUND Primary ovarian insufficiency (POI) refers to the loss of ovarian function under the age of 40 and results in amenorrhea and infertility. Our previous studies have shown that transplantation of mesenchymal stem cells (MSCs) and MSC-derived exosomes in chemotherapy-induced POI mouse ovaries can reverse the POI and eventually achieve pregnancy. Based on our recent studies, MSC-derived exosomes have almost equal therapeutic potentials as transplanted MSCs. However, it is still unclear whether exosomes can completely replace MSCs in POI treatment. For the reliable application of cell-free treatment for POI patients using exosomes, there is a need to understand whether there is any outcome and effectiveness difference between MSC and MSC-derived exosome treatment. METHODS Comparing the therapeutic effect of intravenous injection using MSCs and equal amounts of exosomes in a POI mouse model will reveal the difference between the two therapeutic resources. In this study, we induced POI in C57/BL6 mice by chemotherapy (CXT) using a standard protocol. We then injected four different doses of MSCs or equal amounts of commercialized MSC-derived exosomes by retro-orbital injection post-CXT. RESULT After MSC/exosome treatment, tissue and serum samples were harvested to analyze molecular changes after treatment, while other mice in parallel experiments underwent breeding experiments to compare the restoration of fertility. Both the MSC- and exosome-treated groups had a restored estrous cycle and serum hormone levels compared to untreated POI mice. The pregnancy rate in the MSC-treated group was 60-100% after treatment, while the pregnancy rate in the exosome-treated group was 30-50% after treatment. Interestingly, in terms of long-term effects, MSC-treated mice still showed a 60-80% pregnancy rate in the second round of breeding, while the exosome-treated group became infertile again in the second round of breeding. CONCLUSIONS Although there were some differences in the efficacy between MSC treatment and exosome treatment, both treatments were able to achieve pregnancy in the POI mouse model. In conclusion, we report that MSC-derived exosomes are a promising therapeutic option to restore ovarian function in POI conditions similar to treatment with MSCs.
Collapse
Affiliation(s)
- Hang-Soo Park
- Department of Obstetrics and Gynecology, University of Chicago, 5841 S. Maryland Ave., Chicago, IL, 60637, USA
| | - Rishi Man Chugh
- Department of Surgery, University of Illinois at Chicago, Chicago, IL, 60612, USA
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Jin Seok
- Department of Obstetrics and Gynecology, University of Chicago, 5841 S. Maryland Ave., Chicago, IL, 60637, USA
| | - Esra Cetin
- Department of Obstetrics and Gynecology, University of Chicago, 5841 S. Maryland Ave., Chicago, IL, 60637, USA
| | - Hanaa Mohammed
- Department of Obstetrics and Gynecology, University of Chicago, 5841 S. Maryland Ave., Chicago, IL, 60637, USA
- Human Anatomy and Embryology Department, Faculty of Medicine, Sohag University, Sohag, 82524, Egypt
| | - Hiba Siblini
- Department of Obstetrics and Gynecology, University of Chicago, 5841 S. Maryland Ave., Chicago, IL, 60637, USA
| | - Farzana Liakath Ali
- Department of Obstetrics and Gynecology, University of Chicago, 5841 S. Maryland Ave., Chicago, IL, 60637, USA
| | | | - Hiba Alkelani
- Department of Obstetrics and Gynecology, University of Chicago, 5841 S. Maryland Ave., Chicago, IL, 60637, USA
| | - Amro Elsharoud
- Department of Surgery, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Mara Ulin
- Department of Surgery, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Sahar Esfandyari
- Department of Surgery, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Ayman Al-Hendy
- Department of Obstetrics and Gynecology, University of Chicago, 5841 S. Maryland Ave., Chicago, IL, 60637, USA.
- Department of Surgery, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
14
|
Yuan YG, Wang JL, Zhang YX, Li L, Reza AMMT, Gurunathan S. Biogenesis, Composition and Potential Therapeutic Applications of Mesenchymal Stem Cells Derived Exosomes in Various Diseases. Int J Nanomedicine 2023; 18:3177-3210. [PMID: 37337578 PMCID: PMC10276992 DOI: 10.2147/ijn.s407029] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/31/2023] [Indexed: 06/21/2023] Open
Abstract
Exosomes are nanovesicles with a wide range of chemical compositions used in many different applications. Mesenchymal stem cell-derived exosomes (MSCs-EXOs) are spherical vesicles that have been shown to mediate tissue regeneration in a variety of diseases, including neurological, autoimmune and inflammatory, cancer, ischemic heart disease, lung injury, and liver fibrosis. They can modulate the immune response by interacting with immune effector cells due to the presence of anti-inflammatory compounds and are involved in intercellular communication through various types of cargo. MSCs-EXOs exhibit cytokine storm-mitigating properties in response to COVID-19. This review discussed the potential function of MSCs-EXOs in a variety of diseases including neurological, notably epileptic encephalopathy and Parkinson's disease, cancer, angiogenesis, autoimmune and inflammatory diseases. We provided an overview of exosome biogenesis and factors that regulate exosome biogenesis. Additionally, we highlight the functions and potential use of MSCs-EXOs in the treatment of the inflammatory disease COVID-19. Finally, we covered a strategies and challenges of MSCs-EXOs. Finally, we discuss conclusion and future perspectives of MSCs-EXOs.
Collapse
Affiliation(s)
- Yu-Guo Yuan
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
- Jiangsu Co-Innovation Center of Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
| | - Jia-Lin Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
- Jiangsu Co-Innovation Center of Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
| | - Ya-Xin Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
- Jiangsu Co-Innovation Center of Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
| | - Ling Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
- Jiangsu Co-Innovation Center of Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, People’s Republic of China
| | - Abu Musa Md Talimur Reza
- Department of Molecular Biology and Genetics, Faculty of Science, Gebze Technical University, Gebze, Kocaeli, Türkiye
| | | |
Collapse
|
15
|
Leyfman Y, Gohring G, Joshi M, Menon GP, Van de Kieft A, Rivero TD, Bellio MA, Mitrani MI. Extracellular vesicles: A promising therapy against SARS-CoV-2 infection. Mol Ther 2023; 31:1196-1200. [PMID: 37141856 PMCID: PMC10155280 DOI: 10.1016/j.ymthe.2023.03.033] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 03/31/2023] [Accepted: 03/31/2023] [Indexed: 05/06/2023] Open
Affiliation(s)
- Yan Leyfman
- Icahn School of Medicine at Mount Sinai South Nassau, Oceanside, NY, USA
| | | | - Muskan Joshi
- Tbilisi State Medical University, Tbilisi, Georgia
| | | | | | | | | | | |
Collapse
|
16
|
Leyfman Y, Emmanuel N, Menon GP, Joshi M, Wilkerson WB, Cappelli J, Erick TK, Park CH, Sharma P. Cancer and COVID-19: unravelling the immunological interplay with a review of promising therapies against severe SARS-CoV-2 for cancer patients. J Hematol Oncol 2023; 16:39. [PMID: 37055774 PMCID: PMC10100631 DOI: 10.1186/s13045-023-01432-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/25/2023] [Indexed: 04/15/2023] Open
Abstract
Cancer patients, due to their immunocompromised status, are at an increased risk for severe SARS-CoV-2 infection. Since severe SARS-CoV-2 infection causes multiple organ damage through IL-6-mediated inflammation while stimulating hypoxia, and malignancy promotes hypoxia-induced cellular metabolic alterations leading to cell death, we propose a mechanistic interplay between both conditions that results in an upregulation of IL-6 secretion resulting in enhanced cytokine production and systemic injury. Hypoxia mediated by both conditions results in cell necrosis, dysregulation of oxidative phosphorylation, and mitochondrial dysfunction. This produces free radicals and cytokines that result in systemic inflammatory injury. Hypoxia also catalyzes the breakdown of COX-1 and 2 resulting in bronchoconstriction and pulmonary edema, which further exacerbates tissue hypoxia. Given this disease model, therapeutic options are currently being studied against severe SARS-COV-2. In this study, we review several promising therapies against severe disease supported by clinical trial evidence-including Allocetra, monoclonal antibodies (Tixagevimab-Cilgavimab), peginterferon lambda, Baricitinib, Remdesivir, Sarilumab, Tocilizumab, Anakinra, Bevacizumab, exosomes, and mesenchymal stem cells. Due to the virus's rapid adaptive evolution and diverse symptomatic manifestation, the use of combination therapies offers a promising approach to decrease systemic injury. By investing in such targeted interventions, cases of severe SARS-CoV-2 should decrease along with its associated long-term sequelae and thereby allow cancer patients to resume their treatments.
Collapse
Affiliation(s)
- Yan Leyfman
- Icahn School of Medicine at Mount Sinai South Nassau, Rockville Centre, NY, USA
| | - Nancy Emmanuel
- Hospital das Clínicas of the Faculty of Medicine of the University of São Paulo, São Paulo, Brazil
| | | | - Muskan Joshi
- Tbilisi State Medical University, Tbilisi, Georgia
| | | | | | | | | | - Pushpa Sharma
- Department of Anesthesiology, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| |
Collapse
|
17
|
Ora J, Calzetta L, Frugoni C, Puxeddu E, Rogliani P. Expert guidance on the management and challenges of long-COVID syndrome: a systematic review. Expert Opin Pharmacother 2023; 24:315-330. [PMID: 36542805 DOI: 10.1080/14656566.2022.2161365] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Long-COVID is a condition characterized by the permanence of symptoms beyond 4 weeks after an initial infection. It affects 1 out of 5 people and is loosely related to the severity of acute infection and pathological mechanisms, which are yet to be understood. AREAS COVERED This article looks at currently available and under-studied therapies for long-COVID syndrome. It particularly gives focus to ongoing trials and reviews the underlying mechanisms. A comprehensive literature search was performed on PubMed and clincaltrial.gov of clinical trials concerning the management of long-COVID syndrome. EXPERT OPINION 'Long-COVID' syndrome is a new emergency characterized by several symptoms such as fatigue, dyspnea, cognitive and attention disorders, sleep disorders, post-traumatic stress disorder, muscle pain, and concentration problems. Despite the many guidelines available to date, there are no established treatments of long-COVID. Pharmacological research is studying known drugs that act on the reduction or modulation of systemic inflammation, or innovative drugs used in similar pathologies. Rehabilitation now seems to be the safest treatment to offer, whereas we will have to wait for the pharmacological research trials in progress as well as plan new trials based on a better understanding of the pathogenic mechanisms.
Collapse
Affiliation(s)
- Josuel Ora
- Unit of Respiratory Medicine, Division of Emergency Medicine, University Hospital Tor Vergata, 00133, Rome, Italy.,Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - Luigino Calzetta
- Respiratory Disease and Lung Function Unit, Department of Medicine and Surgery, University of Parma, 43126, Parma, Italy
| | - Chiara Frugoni
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - Ermanno Puxeddu
- Unit of Respiratory Medicine, Division of Emergency Medicine, University Hospital Tor Vergata, 00133, Rome, Italy.,Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - Paola Rogliani
- Unit of Respiratory Medicine, Division of Emergency Medicine, University Hospital Tor Vergata, 00133, Rome, Italy.,Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome "Tor Vergata", 00133, Rome, Italy
| |
Collapse
|
18
|
Nunzi E, Mezzasoma L, Bellezza I, Zelante T, Orvietani P, Coata G, Giardina I, Sagini K, Manni G, Di Michele A, Gargaro M, Talesa VN, Di Renzo GC, Fallarino F, Romani R. Microbiota-Associated HAF-EVs Regulate Monocytes by Triggering or Inhibiting Inflammasome Activation. Int J Mol Sci 2023; 24:ijms24032527. [PMID: 36768851 PMCID: PMC9916438 DOI: 10.3390/ijms24032527] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/21/2023] [Accepted: 01/25/2023] [Indexed: 02/03/2023] Open
Abstract
In pregnancy, human amniotic fluid extracellular vesicles (HAF-EVs) exert anti-inflammatory effects on T cells and on monocytes, supporting their immunoregulatory roles. The specific mechanisms are still not completely defined. The aim of this study was to investigate the ability of HAF-EVs, isolated from pregnant women who underwent amniocentesis and purified by gradient ultracentrifugation, to affect inflammasome activation in the human monocytes. Proteomic studies revealed that HAF-EV samples expressed several immunoregulatory molecules as well as small amounts of endotoxin. Surprisingly, metagenomic analysis shows the presence of specific bacterial strain variants associated with HAF-EVs as potential sources of the endotoxin. Remarkably, we showed that a single treatment of THP-1 cells with HAF-EVs triggered inflammasome activation, whereas the same treatment followed by LPS and ATP sensitization prevented inflammasome activation, a pathway resembling monocyte refractories. A bioinformatics analysis of microbiota-HAF-EVs functional pathways confirmed the presence of enzymes for endotoxin biosynthesis as well as others associated with immunoregulatory functions. Overall, these data suggest that HAF-EVs could serve as a source of the isolation of a specific microbiota during early pregnancy. Moreover, HAF-EVs could act as a novel system to balance immune training and tolerance by modulating the inflammasome in monocytes or other cells.
Collapse
Affiliation(s)
- Emilia Nunzi
- Department of Medicine and Surgery, University of Perugia, Polo Unico Sant’Andrea delle Fratte, P.e Lucio Severi 1, 06132 Perugia, Italy
| | - Letizia Mezzasoma
- Department of Medicine and Surgery, University of Perugia, Polo Unico Sant’Andrea delle Fratte, P.e Lucio Severi 1, 06132 Perugia, Italy
| | - Ilaria Bellezza
- Department of Medicine and Surgery, University of Perugia, Polo Unico Sant’Andrea delle Fratte, P.e Lucio Severi 1, 06132 Perugia, Italy
| | - Teresa Zelante
- Department of Medicine and Surgery, University of Perugia, Polo Unico Sant’Andrea delle Fratte, P.e Lucio Severi 1, 06132 Perugia, Italy
| | - Pierluigi Orvietani
- Department of Medicine and Surgery, University of Perugia, Polo Unico Sant’Andrea delle Fratte, P.e Lucio Severi 1, 06132 Perugia, Italy
| | - Giuliana Coata
- Department of Medicine and Surgery, University of Perugia, Polo Unico Sant’Andrea delle Fratte, P.e Lucio Severi 1, 06132 Perugia, Italy
| | - Irene Giardina
- Department of Obstetrics and Gynecology, University Hospital of Perugia, Sant’Andrea delle Fratte, P.e Lucio Severi 1, 06132 Perugia, Italy
| | - Krizia Sagini
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, 0379 Oslo, Norway
| | - Giorgia Manni
- Department of Medicine and Surgery, University of Perugia, Polo Unico Sant’Andrea delle Fratte, P.e Lucio Severi 1, 06132 Perugia, Italy
| | - Alessandro Di Michele
- Department of Physics and Geology, University of Perugia, Via Pascoli, 06123 Perugia, Italy
| | - Marco Gargaro
- Department of Medicine and Surgery, University of Perugia, Polo Unico Sant’Andrea delle Fratte, P.e Lucio Severi 1, 06132 Perugia, Italy
| | - Vincenzo N. Talesa
- Department of Medicine and Surgery, University of Perugia, Polo Unico Sant’Andrea delle Fratte, P.e Lucio Severi 1, 06132 Perugia, Italy
| | - Gian Carlo Di Renzo
- Department of Obstetrics, Gynecology and Perinatology IM Sechenov First State University, 117997 Moscow, Russia
| | - Francesca Fallarino
- Department of Medicine and Surgery, University of Perugia, Polo Unico Sant’Andrea delle Fratte, P.e Lucio Severi 1, 06132 Perugia, Italy
- Correspondence: (F.F.); (R.R.)
| | - Rita Romani
- Department of Medicine and Surgery, University of Perugia, Polo Unico Sant’Andrea delle Fratte, P.e Lucio Severi 1, 06132 Perugia, Italy
- Correspondence: (F.F.); (R.R.)
| |
Collapse
|
19
|
del Rivero T, Milberg J, Bennett C, Mitrani MI, Bellio MA. Human amniotic fluid derived extracellular vesicles attenuate T cell immune response. Front Immunol 2022; 13:977809. [PMID: 36518766 PMCID: PMC9742275 DOI: 10.3389/fimmu.2022.977809] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 11/10/2022] [Indexed: 11/29/2022] Open
Abstract
Introduction Extracellular vesicles isolated from human amniotic fluid (AF-EVs) have previously been found to modulate inflammation and macrophage infiltration in a mouse model. However, the effects of acellular amniotic fluid (acAF) or AF-EVs on the T-Cell immune response have not been explored. Methods In this study, we investigated the effects of acAF and AF-EVs on the T cell immune response in an in vitro cell culture model. Peripheral Blood Mononuclear Cells (PBMCs) were stimulated with Phytohemagglutinin (PHA) to induce the immune response and were subsequently treated with either serum-free media (vehicle), acAF, or concentrated AF-EVs. Results Both acAF and AF-EV treatment suppressed PHA-induced T cell proliferation and PHA-induced T cell activation; however, treatment with concentrated AF-EVs had a greater effect. Additionally, both acAF and AF-EVs reduced PBMC pro-inflammatory cytokine release. AF-EVs were found to be taken up by both CD4+ and CD8+ effector T cell subsets. Conclusion Overall, this data demonstrates that AF-EVs have a robust immunomodulatory effect on T cells and suggests AF-EVs could be used as an immunotherapeutic tool.
Collapse
|
20
|
Tenchov R, Sasso JM, Wang X, Liaw WS, Chen CA, Zhou QA. Exosomes─Nature's Lipid Nanoparticles, a Rising Star in Drug Delivery and Diagnostics. ACS NANO 2022; 16:17802-17846. [PMID: 36354238 PMCID: PMC9706680 DOI: 10.1021/acsnano.2c08774] [Citation(s) in RCA: 252] [Impact Index Per Article: 84.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/21/2022] [Indexed: 05/03/2023]
Abstract
Exosomes are a subgroup of nanosized extracellular vesicles enclosed by a lipid bilayer membrane and secreted by most eukaryotic cells. They represent a route of intercellular communication and participate in a wide variety of physiological and pathological processes. The biological roles of exosomes rely on their bioactive cargos, including proteins, nucleic acids, and lipids, which are delivered to target cells. Their distinctive properties─innate stability, low immunogenicity, biocompatibility, and good biomembrane penetration capacity─allow them to function as superior natural nanocarriers for efficient drug delivery. Another notably favorable clinical application of exosomes is in diagnostics. They hold various biomolecules from host cells, which are indicative of pathophysiological conditions; therefore, they are considered vital for biomarker discovery in clinical diagnostics. Here, we use data from the CAS Content Collection and provide a landscape overview of the current state and delineate trends in research advancement on exosome applications in therapeutics and diagnostics across time, geography, composition, cargo loading, and development pipelines. We discuss exosome composition and pathway, from their biogenesis and secretion from host cells to recipient cell uptake. We assess methods for exosome isolation and purification, their clinical applications in therapy and diagnostics, their development pipelines, the exploration goals of the companies, the assortment of diseases they aim to treat, development stages of their research, and publication trends. We hope this review will be useful for understanding the current knowledge in the field of medical applications of exosomes, in an effort to further solve the remaining challenges in fulfilling their potential.
Collapse
Affiliation(s)
- Rumiana Tenchov
- CAS, a Division of the American Chemical
Society, 2540 Olentangy River Rd, Columbus, Ohio 43202, United
States
| | - Janet M. Sasso
- CAS, a Division of the American Chemical
Society, 2540 Olentangy River Rd, Columbus, Ohio 43202, United
States
| | - Xinmei Wang
- CAS, a Division of the American Chemical
Society, 2540 Olentangy River Rd, Columbus, Ohio 43202, United
States
| | - Wen-Shing Liaw
- CAS, a Division of the American Chemical
Society, 2540 Olentangy River Rd, Columbus, Ohio 43202, United
States
| | - Chun-An Chen
- CAS, a Division of the American Chemical
Society, 2540 Olentangy River Rd, Columbus, Ohio 43202, United
States
| | - Qiongqiong Angela Zhou
- CAS, a Division of the American Chemical
Society, 2540 Olentangy River Rd, Columbus, Ohio 43202, United
States
| |
Collapse
|
21
|
Bowen CM, Ditmars FS, Gupta A, Reems JA, Fagg WS. Cell-Free Amniotic Fluid and Regenerative Medicine: Current Applications and Future Opportunities. Biomedicines 2022; 10:2960. [PMID: 36428527 PMCID: PMC9687956 DOI: 10.3390/biomedicines10112960] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/09/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
Amniotic fluid (AF) provides critical biological and physical support for the developing fetus. While AF is an excellent source of progenitor cells with regenerative properties, recent investigations indicate that cell-free AF (cfAF), which consists of its soluble components and extracellular vesicles, can also stimulate regenerative and reparative activities. This review summarizes published fundamental, translational, and clinical investigations into the biological activity and potential use of cfAF as a therapeutic agent. Recurring themes emerge from these studies, which indicate that cfAF can confer immunomodulatory, anti-inflammatory, and pro-growth characteristics to the target cells/tissue with which they come into contact. Another common observation is that cfAF seems to promote a return of cells/tissue to a homeostatic resting state when applied to a model of cell stress or disease. The precise mechanisms through which these effects are mediated have not been entirely defined, but it is clear that cfAF can safely and effectively treat cutaneous wounds and perhaps orthopedic degenerative conditions. Additional applications are currently being investigated, but require further study to dissect the fundamental mechanisms through which its regenerative effects are mediated. By doing so, rational design can be used to fully unlock its potential in the biotechnology lab and in the clinic.
Collapse
Affiliation(s)
- Charles M. Bowen
- Department of Surgery, The University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
- John Sealy School of Medicine, The University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - Frederick S. Ditmars
- Department of Surgery, The University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
- John Sealy School of Medicine, The University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - Ashim Gupta
- Future Biologics, Lawrenceville, GA 30043, USA
- BioIntegrate, Lawrenceville, GA 30043, USA
- South Texas Orthopaedic Research Institute (STORI Inc.), Laredo, TX 78045, USA
- Regenerative Orthopaedics, Noida 201301, UP, India
| | - Jo-Anna Reems
- Merakris Therapeutics, RTP Frontier 800 Park Offices Dr. Suite 3322, Research Triangle Park, NC 27709, USA
- Department of Engineering, University of Utah, Salt Lake City, UT 84112, USA
| | - William Samuel Fagg
- Department of Surgery, The University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
- Regenerative Orthopaedics, Noida 201301, UP, India
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| |
Collapse
|
22
|
Askenase PW. Recommendation: Treatment of clinical long COVID encephalopathies with nasal administered mesenchymal stromal cell extracellular vesicles. FRONTIERS IN NANOTECHNOLOGY 2022; 4. [DOI: 10.3389/fnano.2022.987117] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
We propose therapy with extracellular vesicles (EVs) for dominant central nervous system aspects of chronic Long COVID Syndromes (LCS). These clinical conditions have a delayed onset of 1–3 months following the cessation of active SARS-CoV-2 virus infections that cause an acute disease called COVID-19. The therapy of LCS will be achieved by direct access to the central nervous system (CNS) by nasal administration of small EVs derived from Mesenchymal Stromal Cells (MSC). When administered nasally, they target CNS microglia and endothelia involved in LCS encephalopathy, as indicated by experimental animal models and human autopsy and spinal fluid studies. Underlying this approach is the discovery that MSC-sEV treatment for healing neuro injury targets, microglia, and macrophages that then likely release secondary trophic EVs that affect the local capillary endothelial cells to restore vascular integrity. It is postulated that the pathways of endothelial and neural pathologies in acute SARS-CoV-2 virus infections may carry over to produce underlying vascular and neurological defects mediating LCS that are susceptible to this proposed nasal therapy with MSC-sEVs.
Collapse
|
23
|
Mustajab T, Kwamboka MS, Choi DA, Kang DW, Kim J, Han KR, Han Y, Lee S, Song D, Chwae YJ. Update on Extracellular Vesicle-Based Vaccines and Therapeutics to Combat COVID-19. Int J Mol Sci 2022; 23:ijms231911247. [PMID: 36232549 PMCID: PMC9569487 DOI: 10.3390/ijms231911247] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/20/2022] [Accepted: 09/20/2022] [Indexed: 11/23/2022] Open
Abstract
The COVID-19 pandemic has had a deep impact on people worldwide since late 2019 when SARS-CoV-2 was first identified in Wuhan, China. In addition to its effect on public health, it has affected humans in various aspects of life, including social, economic, cultural, and political. It is also true that researchers have made vigorous efforts to overcome COVID-19 throughout the world, but they still have a long way to go. Accordingly, innumerable therapeutics and vaccine candidates have been studied for their efficacies and have been tried clinically in a very short span of time. For example, the versatility of extracellular vesicles, which are membrane-bound particles released from all types of cells, have recently been highlighted in terms of their effectiveness, biocompatibility, and safety in the fight against COVID-19. Thus, here, we tried to explain the use of extracellular vesicles as therapeutics and for the development of vaccines against COVID-19. Along with the mechanisms and a comprehensive background of their application in trapping the coronavirus or controlling the cytokine storm, we also discuss the obstacles to the clinical use of extracellular vesicles and how these could be resolved in the future.
Collapse
Affiliation(s)
- Tamanna Mustajab
- Department of Microbiology, School of Medicine, Ajou University, Suwon 16499, Korea
- Department of Biomedical Science, Graduate School of Ajou University, Suwon 16499, Korea
| | - Moriasi Sheba Kwamboka
- Department of Microbiology, School of Medicine, Ajou University, Suwon 16499, Korea
- Department of Biomedical Science, Graduate School of Ajou University, Suwon 16499, Korea
| | - Da Ae Choi
- Department of Microbiology, School of Medicine, Ajou University, Suwon 16499, Korea
- Department of Biomedical Science, Graduate School of Ajou University, Suwon 16499, Korea
| | - Dae Wook Kang
- Department of Microbiology, School of Medicine, Ajou University, Suwon 16499, Korea
- Department of Biomedical Science, Graduate School of Ajou University, Suwon 16499, Korea
| | - Junho Kim
- Department of Microbiology, School of Medicine, Ajou University, Suwon 16499, Korea
- Department of Biomedical Science, Graduate School of Ajou University, Suwon 16499, Korea
| | - Kyu Ri Han
- Department of Microbiology, School of Medicine, Ajou University, Suwon 16499, Korea
- Department of Biomedical Science, Graduate School of Ajou University, Suwon 16499, Korea
| | - Yujin Han
- Department of Microbiology, School of Medicine, Ajou University, Suwon 16499, Korea
- Department of Biomedical Science, Graduate School of Ajou University, Suwon 16499, Korea
| | - Sorim Lee
- Department of Microbiology, School of Medicine, Ajou University, Suwon 16499, Korea
- Department of Biomedical Science, Graduate School of Ajou University, Suwon 16499, Korea
| | - Dajung Song
- Department of Microbiology, School of Medicine, Ajou University, Suwon 16499, Korea
- Department of Biomedical Science, Graduate School of Ajou University, Suwon 16499, Korea
| | - Yong-Joon Chwae
- Department of Microbiology, School of Medicine, Ajou University, Suwon 16499, Korea
- Department of Biomedical Science, Graduate School of Ajou University, Suwon 16499, Korea
- Correspondence: ; Tel.: +82-031-219-5073
| |
Collapse
|
24
|
Liu C, Xiao K, Xie L. Advances in the use of exosomes for the treatment of ALI/ARDS. Front Immunol 2022; 13:971189. [PMID: 36016948 PMCID: PMC9396740 DOI: 10.3389/fimmu.2022.971189] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/22/2022] [Indexed: 11/16/2022] Open
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a critical clinical syndrome with high morbidity and mortality. Currently, the primary treatment for ALI/ARDS is mainly symptomatic therapy such as mechanical ventilation and fluid management. Due to the lack of effective treatment strategies, most ALI/ARDS patients face a poor prognosis. The discovery of exosomes has created a promising prospect for the treatment of ALI/ARDS. Exosomes can exert anti-inflammatory effects, inhibit apoptosis, and promote cell regeneration. The microRNA contained in exosomes can participate in intercellular communication and play an immunomodulatory role in ALI/ARDS disease models. This review discusses the possible mechanisms of exosomes in ALI/ARDS to facilitate the development of innovative treatments for ALI/ARDS.
Collapse
Affiliation(s)
- Chang Liu
- School of Medicine, Nankai University, Tianjin, China
- Center of Pulmonary & Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China
| | - Kun Xiao
- Center of Pulmonary & Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China
| | - Lixin Xie
- School of Medicine, Nankai University, Tianjin, China
- Center of Pulmonary & Critical Care Medicine, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China
- *Correspondence: Lixin Xie,
| |
Collapse
|
25
|
Krishnan A, Muthusamy S, Fernandez FB, Kasoju N. Mesenchymal Stem Cell-Derived Extracellular Vesicles in the Management of COVID19-Associated Lung Injury: A Review on Publications, Clinical Trials and Patent Landscape. Tissue Eng Regen Med 2022; 19:659-673. [PMID: 35384633 PMCID: PMC8985390 DOI: 10.1007/s13770-022-00441-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/27/2022] [Accepted: 02/02/2022] [Indexed: 02/07/2023] Open
Abstract
The unprecedented COVID-19 pandemic situation forced the scientific community to explore all the possibilities from various fields, and so far we have seen a lot of surprises, eureka moments and disappointments. One of the approaches from the cellular therapists was exploiting the immunomodulatory and regenerative potential of mesenchymal stromal cells (MSCs), more so of MSC-derived extracellular vesicles (EVs)-particularly exosomes, in order to alleviate the cytokine storm and regenerate the damaged lung tissues. Unlike MSCs, the EVs are easier to store, deliver, and are previously shown to be as effective as MSCs, yet less immunogenic. These features attracted the attention of many and thus led to a tremendous increase in publications, clinical trials and patent applications. This review presents the current landscape of the field and highlights some interesting findings on MSC-derived EVs in the context of COVID-19, including in silico, in vitro, in vivo and case reports. The data strongly suggests the potential of MSC-derived EVs as a therapeutic regime for the management of acute lung injury and associated complications in COVID-19 and beyond.
Collapse
Affiliation(s)
- Anand Krishnan
- Department of Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Science and Technology, Thiruvananthapuram, 695012, Kerala, India
| | - Senthilkumar Muthusamy
- Department of Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Science and Technology, Thiruvananthapuram, 695012, Kerala, India
| | - Francis B Fernandez
- Department of Biomaterial Science and Technology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Science and Technology, Thiruvananthapuram, 695012, Kerala, India
| | - Naresh Kasoju
- Department of Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Science and Technology, Thiruvananthapuram, 695012, Kerala, India.
| |
Collapse
|
26
|
Ruggiero V, Aquino RP, Del Gaudio P, Campiglia P, Russo P. Post-COVID Syndrome: The Research Progress in the Treatment of Pulmonary sequelae after COVID-19 Infection. Pharmaceutics 2022; 14:pharmaceutics14061135. [PMID: 35745708 PMCID: PMC9229559 DOI: 10.3390/pharmaceutics14061135] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/20/2022] [Accepted: 05/25/2022] [Indexed: 12/15/2022] Open
Abstract
Post-COVID syndrome or long COVID is defined as the persistence of symptoms after confirmed SARS-CoV-2 infection, the pathogen responsible for coronavirus disease. The content herein presented reviews the reported long-term consequences and aftereffects of COVID-19 infection and the potential strategies to adopt for their management. Recent studies have shown that severe forms of COVID-19 can progress into acute respiratory distress syndrome (ARDS), a predisposing factor of pulmonary fibrosis that can irreversibly compromise respiratory function. Considering that the most serious complications are observed in the airways, the inhalation delivery of drugs directly to the lungs should be preferred, since it allows to lower the dose and systemic side effects. Although further studies are needed to optimize these techniques, recent studies have also shown the importance of in vitro models to recreate the SARS-CoV-2 infection and study its sequelae. The information reported suggests the necessity to develop new inhalation therapies in order to improve the quality of life of patients who suffer from this condition.
Collapse
Affiliation(s)
- Valentina Ruggiero
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (V.R.); (R.P.A.); (P.D.G.); (P.C.)
- PhD Program in Drug Discovery and Development, University of Salerno, 84084 Fisciano, Italy
| | - Rita P. Aquino
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (V.R.); (R.P.A.); (P.D.G.); (P.C.)
| | - Pasquale Del Gaudio
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (V.R.); (R.P.A.); (P.D.G.); (P.C.)
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (V.R.); (R.P.A.); (P.D.G.); (P.C.)
| | - Paola Russo
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy; (V.R.); (R.P.A.); (P.D.G.); (P.C.)
- Correspondence:
| |
Collapse
|
27
|
Gebara N, Scheel J, Skovronova R, Grange C, Marozio L, Gupta S, Giorgione V, Caicci F, Benedetto C, Khalil A, Bussolati B. Single extracellular vesicle analysis in human amniotic fluid shows evidence of phenotype alterations in preeclampsia. J Extracell Vesicles 2022; 11:e12217. [PMID: 35582873 PMCID: PMC9115584 DOI: 10.1002/jev2.12217] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 02/24/2022] [Accepted: 03/29/2022] [Indexed: 12/14/2022] Open
Abstract
Amniotic fluid surrounding the developing fetus is a complex biological fluid rich in metabolically active bio-factors. The presence of extracellular vesicles (EVs) in amniotic fluid has been mainly related to foetal urine. We here characterized EVs from term amniotic fluid in terms of surface marker expression using different orthogonal techniques. EVs appeared to be a heterogeneous population expressing markers of renal, placental, epithelial and stem cells. Moreover, we compared amniotic fluid EVs from normal pregnancies with those of preeclampsia, a hypertensive disorder affecting up to 8% of pregnancies worldwide. An increase of CD105 (endoglin) expressing EVs was observed in preeclamptic amniotic fluid by bead-based cytofluorimetric analysis, and further confirmed using a chip-based analysis. HLA-G, a typical placental marker, was not co-expressed by the majority of CD105+ EVs, in analogy with amniotic fluid stromal cell derived-EVs. At a functional level, preeclampsia-derived EVs, but not normal pregnancy EVs, showed an antiangiogenic effect, possibly due to the decoy effect of endoglin. Our results provide a characterization of term amniotic fluid-EVs, supporting their origin from foetal and placental cells. In preeclampsia, the observed antiangiogenic characteristics of amniotic fluid-EVs may reflect the hypoxic and antiangiogenic microenvironment and could possibly impact on the developing fetus or on the surrounding foetal membranes.
Collapse
Affiliation(s)
- Natalia Gebara
- Department of Molecular Biotechnology and Health SciencesUniversity of TurinTurinItaly
| | - Julia Scheel
- Department of Systems Biology and BioinformaticsUniversity of RostockRostockGermany
| | - Renata Skovronova
- Department of Molecular Biotechnology and Health SciencesUniversity of TurinTurinItaly
| | | | - Luca Marozio
- Department of Surgical Sciences, Obstetrics and Gynecology, University of TurinTurinItaly
| | - Shailendra Gupta
- Department of Systems Biology and BioinformaticsUniversity of RostockRostockGermany
| | - Veronica Giorgione
- Vascular Biology Research CentreMolecular and Clinical Sciences Research InstituteSt George's University of LondonLondonUK
| | | | - Chiara Benedetto
- Department of Surgical Sciences, Obstetrics and Gynecology, University of TurinTurinItaly
| | - Asma Khalil
- Vascular Biology Research CentreMolecular and Clinical Sciences Research InstituteSt George's University of LondonLondonUK
- Foetal Medicine UnitSt George's University Hospitals NHS Foundation TrustSt George's University of LondonLondonUK
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health SciencesUniversity of TurinTurinItaly
| |
Collapse
|
28
|
Kaffash Farkhad N, Mahmoudi A, Mahdipour E. Regenerative therapy by using Mesenchymal Stem Cells-derived exosomes in COVID-19 treatment. The potential role and underlying mechanisms. Regen Ther 2022; 20:61-71. [PMID: 35340407 PMCID: PMC8938276 DOI: 10.1016/j.reth.2022.03.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/04/2022] [Accepted: 03/13/2022] [Indexed: 12/03/2022] Open
Abstract
COVID-19 disease caused by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), started in December 2019 in Wuhan, China, and quickly became the global pandemic. The high spread rate, relatively high mortality rate, and the lack of specific medicine have led researchers and clinicians worldwide to find new treatment strategies. Unfortunately, evidence shows that the virus-specific receptor Angiotensin-Converting Enzyme 2 (ACE-2) is present on the surface of most cells in the body, leading to immune system dysfunction and multi-organ failure in critically ill patients. In this context, the use of Mesenchymal Stem Cells (MSCs) and their secret has opened new therapeutic horizons for patients due to the lack of ACE2 receptor expression. MSCs exert their beneficial therapeutic actions, particularly anti-inflammatory and immunomodulatory properties, mainly through paracrine effects which are mediated by exosomes. Exosomes are bilayer nanovesicles that carry a unique cargo of proteins, lipids and functional nucleic acids based on their cell origin. This review article aims to investigate the possible role of exosomes and the underlying mechanism involved in treating COVID-19 disease based on recent findings.
Collapse
Affiliation(s)
- Najmeh Kaffash Farkhad
- Immunology Research Center, Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Mahmoudi
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elahe Mahdipour
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Corresponding author. Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, University campus. Azadi Sq, Mashhad. Iran.
| |
Collapse
|
29
|
Loo CY, Lee WH. Nanotechnology-based therapeutics for targeting inflammatory lung diseases. Nanomedicine (Lond) 2022; 17:865-879. [PMID: 35315290 DOI: 10.2217/nnm-2021-0447] [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: 11/21/2022] Open
Abstract
The physiochemical properties of drugs used in treating inflammation-associated lung diseases (i.e., asthma, chronic obstructive pulmonary disease, pulmonary fibrosis) play an important role in determining the effectiveness of formulations. Most commonly used drugs are associated with low solubility, low stability and rapid clearance, thus resulting in low bioavailability and therapeutic index. This review focuses on current trends and development of drugs (i.e., corticosteroids, long-acting β-agonists and biomacromolecules such as DNA, siRNA and mRNA) employed to treat inflammatory lung diseases. In addition, this review includes the current challenges of and future perspective with regard to nanotechnology in the treatment of inflammatory lung diseases.
Collapse
Affiliation(s)
- Ching-Yee Loo
- Faculty of Pharmacy and Health Sciences, Royal College of Medicine Perak, Universiti Kuala Lumpur, Ipoh, Perak, 30450, Malaysia
| | - Wing-Hin Lee
- Faculty of Pharmacy and Health Sciences, Royal College of Medicine Perak, Universiti Kuala Lumpur, Ipoh, Perak, 30450, Malaysia
| |
Collapse
|
30
|
Thomas SC, Kim JW, Pauletti GM, Hassett DJ, Kotagiri N. Exosomes: Biological Pharmaceutical Nanovectors for Theranostics. Front Bioeng Biotechnol 2022; 9:808614. [PMID: 35096795 PMCID: PMC8790084 DOI: 10.3389/fbioe.2021.808614] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 12/24/2021] [Indexed: 12/12/2022] Open
Abstract
Exosomes are natural cell-derived nanovesicles of endocytic origin that enable cellular crosstalk by transferring encapsulated molecular cargos across biological barriers, thereby holding significantly complex implications in the etiology and progression of diverse disease states. Consequently, the development of exosomes-based nano-theranostic strategies has received immense consideration for advancing therapeutic interventions and disease prognosis. Their favorable biopharmaceutical properties make exosomes a unique nanoparticulate carrier for pharmaceutical drug delivery. This review provides an update on the contemporary strategies utilizing exosomes for theranostic applications in nanomedicine. In addition, we provide a synopsis of exosomal features and insights into strategic modifications that control in vivo biodistribution. We further discuss their opportunities, merits and pitfalls for cell/tissue targeted drug delivery in personalized nanotherapy.
Collapse
Affiliation(s)
- Shindu C Thomas
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, United States
| | - Jin-Woo Kim
- Department of Biological and Agricultural Engineering, Institute for Nanoscience and Engineering, University of Arkansas, Fayetteville, AR, United States
| | - Giovanni M Pauletti
- St. Louis College of Pharmacy, University of Health Sciences and Pharmacy in St. Louis, St. Louis, MO, United States
| | - Daniel J Hassett
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Nalinikanth Kotagiri
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
31
|
Costa A, Quarto R, Bollini S. Small Extracellular Vesicles from Human Amniotic Fluid Samples as Promising Theranostics. Int J Mol Sci 2022; 23:ijms23020590. [PMID: 35054775 PMCID: PMC8775841 DOI: 10.3390/ijms23020590] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/02/2022] [Accepted: 01/04/2022] [Indexed: 02/05/2023] Open
Abstract
Since the first evidence that stem cells can provide pro-resolving effects via paracrine secretion of soluble factors, growing interest has been addressed to define the most ideal cell source for clinical translation. Leftover or clinical waste samples of human amniotic fluid obtained following prenatal screening, clinical intervention, or during scheduled caesarean section (C-section) delivery at term have been recently considered an appealing source of mesenchymal progenitors with peculiar regenerative capacity. Human amniotic fluid stem cells (hAFSC) have been demonstrated to support tissue recovery in several preclinical models of disease by exerting paracrine proliferative, anti-inflammatory and regenerative influence. Small extracellular vesicles (EVs) concentrated from the hAFSC secretome (the total soluble trophic factors secreted in the cell-conditioned medium, hAFSC-CM) recapitulate most of the beneficial cell effects. Independent studies in preclinical models of either adult disorders or severe diseases in newborns have suggested a regenerative role of hAFSC-EVs. EVs can be eventually concentrated from amniotic fluid (hAF) to offer useful prenatal information, as recently suggested. In this review, we focus on the most significant aspects of EVs obtained from either hAFSC and hAF and consider the current challenges for their clinical translation, including isolation, characterization and quantification methods.
Collapse
Affiliation(s)
- Ambra Costa
- Experimental Biology Unit, Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (A.C.); (R.Q.)
| | - Rodolfo Quarto
- Experimental Biology Unit, Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (A.C.); (R.Q.)
- Cellular Oncology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Sveva Bollini
- Experimental Biology Unit, Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (A.C.); (R.Q.)
- Correspondence: ; Tel.: +39-010-555-8394
| |
Collapse
|
32
|
Bellio MA, Bennett C, Arango A, Khan A, Xu X, Barrera C, Friedewald V, Mitrani MI. Proof-of-concept trial of an amniotic fluid-derived extracellular vesicle biologic for treating high risk patients with mild-to-moderate acute COVID-19 infection. BIOMATERIALS AND BIOSYSTEMS 2021; 4:100031. [PMID: 34841370 PMCID: PMC8611818 DOI: 10.1016/j.bbiosy.2021.100031] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/11/2021] [Accepted: 11/21/2021] [Indexed: 12/09/2022] Open
Abstract
A pandemic brought on by COVID-19 has created a scalable health crisis. The search to help alleviate COVID-19-related complications through therapeutics has become a necessity. Zofin is an investigational, acellular biologic derived from full-term perinatal amniotic fluid that contains extracellular vesicles. Extracellular nanoparticles as such have been studied for their immunomodulatory benefits via cellular therapeutics and, if applied to COVID-19-related inflammation, could benefit patient outcome. Subjects (n = 8) experiencing mild-to-moderate COVID-19 symptoms were treated with the experimental intervention. Complete blood count, complete metabolic panel, inflammatory biomarkers, and absolute lymphocyte counts were recorded prior to and on days 4, 8, 14, 21, and 30 as markers of disease progression. Additionally, chest x-rays were taken of the patients prior to and on days 8 and 30. Patients experienced no serious adverse events. All COVID-19-associated symptoms resolved or became stable with no indication of disease worsening as found by patient and chest x-ray reports. Inflammatory biomarkers (CRP, IL-6, TNF- α ) and absolute lymphocyte counts improved throughout the study period. Findings from a proof-of-concept, expanded access trial for COVID-19 patients prove the acellular biologic is safe and potentially effective to prevent disease progression in a high-risk COVID-19 population with mild-to-moderate symptoms.
Collapse
Affiliation(s)
| | - Cassie Bennett
- Organicell Regenerative Medicine, Miami, FL 33136, United States
| | - Alissa Arango
- Organicell Regenerative Medicine, Miami, FL 33136, United States
| | - Aisha Khan
- Assure Immune LLC., Miami, FL 33136, United States
| | - Xiumin Xu
- Assure Immune LLC., Miami, FL 33136, United States
| | - Cesar Barrera
- United Memorial Medical Center, Houston, TX 77091, United States
| | | | - Maria Ines Mitrani
- Organicell Regenerative Medicine, Miami, FL 33136, United States,Corresponding author at: Organicell Regenerative Medicine, Inc. 1951 Northwest 7th Ave, Suite #300, Miami, FL 33136, United States.
| |
Collapse
|
33
|
Abbas M, Alqahtani MS, Almohiy HM, Alqahtani FF, Alhifzi R, Jambi LK. The Potential Contribution of Biopolymeric Particles in Lung Tissue Regeneration of COVID-19 Patients. Polymers (Basel) 2021; 13:4011. [PMID: 34833310 PMCID: PMC8623030 DOI: 10.3390/polym13224011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/12/2021] [Accepted: 11/16/2021] [Indexed: 02/08/2023] Open
Abstract
The lung is a vital organ that houses the alveoli, which is where gas exchange takes place. The COVID-19 illness attacks lung cells directly, creating significant inflammation and resulting in their inability to function. To return to the nature of their job, it may be essential to rejuvenate the afflicted lung cells. This is difficult because lung cells need a long time to rebuild and resume their function. Biopolymeric particles are the most effective means to transfer developing treatments to airway epithelial cells and then regenerate infected lung cells, which is one of the most significant symptoms connected with COVID-19. Delivering biocompatible and degradable natural biological materials, chemotherapeutic drugs, vaccines, proteins, antibodies, nucleic acids, and diagnostic agents are all examples of these molecules' usage. Furthermore, they are created by using several structural components, which allows them to effectively connect with these cells. We highlight their most recent uses in lung tissue regeneration in this review. These particles are classified into three groups: biopolymeric nanoparticles, biopolymeric stem cell materials, and biopolymeric scaffolds. The techniques and processes for regenerating lung tissue will be thoroughly explored.
Collapse
Affiliation(s)
- Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha 61421, Saudi Arabia
- Computers and Communications Department, College of Engineering, Delta University for Science and Technology, Gamasa 35712, Egypt
| | - Mohammed S. Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia; (M.S.A.); (H.M.A.); (R.A.)
- BioImaging Unit, Space Research Centre, Michael Atiyah Building, University of Leicester, Leicester LE1 7RH, UK
| | - Hussain M. Almohiy
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia; (M.S.A.); (H.M.A.); (R.A.)
| | - Fawaz F. Alqahtani
- Department of Radiological Sciences, College of Applied Medical Sciences, Najran University, Najran 1988, Saudi Arabia;
| | - Roaa Alhifzi
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia; (M.S.A.); (H.M.A.); (R.A.)
| | - Layal K. Jambi
- Radiological Sciences Department, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh 11433, Saudi Arabia;
| |
Collapse
|
34
|
Claridge B, Lozano J, Poh QH, Greening DW. Development of Extracellular Vesicle Therapeutics: Challenges, Considerations, and Opportunities. Front Cell Dev Biol 2021; 9:734720. [PMID: 34616741 PMCID: PMC8488228 DOI: 10.3389/fcell.2021.734720] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 07/30/2021] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) hold great promise as therapeutic modalities due to their endogenous characteristics, however, further bioengineering refinement is required to address clinical and commercial limitations. Clinical applications of EV-based therapeutics are being trialed in immunomodulation, tissue regeneration and recovery, and as delivery vectors for combination therapies. Native/biological EVs possess diverse endogenous properties that offer stability and facilitate crossing of biological barriers for delivery of molecular cargo to cells, acting as a form of intercellular communication to regulate function and phenotype. Moreover, EVs are important components of paracrine signaling in stem/progenitor cell-based therapies, are employed as standalone therapies, and can be used as a drug delivery system. Despite remarkable utility of native/biological EVs, they can be improved using bio/engineering approaches to further therapeutic potential. EVs can be engineered to harbor specific pharmaceutical content, enhance their stability, and modify surface epitopes for improved tropism and targeting to cells and tissues in vivo. Limitations currently challenging the full realization of their therapeutic utility include scalability and standardization of generation, molecular characterization for design and regulation, therapeutic potency assessment, and targeted delivery. The fields' utilization of advanced technologies (imaging, quantitative analyses, multi-omics, labeling/live-cell reporters), and utility of biocompatible natural sources for producing EVs (plants, bacteria, milk) will play an important role in overcoming these limitations. Advancements in EV engineering methodologies and design will facilitate the development of EV-based therapeutics, revolutionizing the current pharmaceutical landscape.
Collapse
Affiliation(s)
- Bethany Claridge
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Melbourne, VIC, Australia
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Jonathan Lozano
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC, Australia
| | - Qi Hui Poh
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Melbourne, VIC, Australia
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - David W. Greening
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Melbourne, VIC, Australia
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Central Clinical School, Monash University, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
35
|
Mitrani MI, Bellio MA, Meglin A, Khan A, Xu X, Haskell G, Arango A, Shapiro GC. Treatment of a COVID-19 long hauler with an amniotic fluid-derived extracellular vesicle biologic. Respir Med Case Rep 2021; 34:101502. [PMID: 34485048 PMCID: PMC8405236 DOI: 10.1016/j.rmcr.2021.101502] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/16/2021] [Accepted: 08/24/2021] [Indexed: 12/24/2022] Open
Abstract
Post-COVID-19 infection symptoms such as mental fog, tachycardia, and extreme fatigue are just a few of the symptoms wreaking havoc on patients’ lives. Patients with long-term symptoms following COVID-19 are being called long haulers. To date, long haulers are receiving little to no guidance from physicians on their lingering COVID-19 symptoms with limited treatment options available. Zofin is an acellular biologic that contains the extracellular vesicle (EV) fraction of human amniotic fluid and is under investigation for use as a COVID-19 therapeutic. We obtained FDA and IRB approval to investigate the therapeutic use of Zofin in a single long hauler patient case experiencing prolonged shortness of breath and respiratory impairment. Administration of the EV product was shown to be safe. Furthermore, demonstrated respiratory improvements through chest X ray images and oxygen saturation measurement. The single patient IND studies were completed without any reported adverse events or safety concerns. Furthermore, these completed studies demonstrate the feasibility and a therapeutic potential of amniotic fluid-derived EVs for COVID-19 long hauler intervention.
Collapse
Affiliation(s)
| | | | - Allen Meglin
- Advanced Regenerative Therapies, Savanah, GA, USA
| | | | | | | | - Alissa Arango
- Organicell Regenerative Medicine, Inc., Miami, FL, USA
| | | |
Collapse
|
36
|
Mosquera-Heredia MI, Morales LC, Vidal OM, Barceló E, Silvera-Redondo C, Vélez JI, Garavito-Galofre P. Exosomes: Potential Disease Biomarkers and New Therapeutic Targets. Biomedicines 2021; 9:1061. [PMID: 34440265 PMCID: PMC8393483 DOI: 10.3390/biomedicines9081061] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 02/07/2023] Open
Abstract
Exosomes are extracellular vesicles released by cells, both constitutively and after cell activation, and are present in different types of biological fluid. Exosomes are involved in the pathogenesis of diseases, such as cancer, neurodegenerative diseases, pregnancy disorders and cardiovascular diseases, and have emerged as potential non-invasive biomarkers for the detection, prognosis and therapeutics of a myriad of diseases. In this review, we describe recent advances related to the regulatory mechanisms of exosome biogenesis, release and molecular composition, as well as their role in health and disease, and their potential use as disease biomarkers and therapeutic targets. In addition, the advantages and disadvantages of their main isolation methods, characterization and cargo analysis, as well as the experimental methods used for exosome-mediated drug delivery, are discussed. Finally, we present potential perspectives for the use of exosomes in future clinical practice.
Collapse
Affiliation(s)
- Maria I. Mosquera-Heredia
- Department of Medicine, Universidad del Norte, Barranquilla 081007, Colombia; (L.C.M.); (O.M.V.); (C.S.-R.)
| | - Luis C. Morales
- Department of Medicine, Universidad del Norte, Barranquilla 081007, Colombia; (L.C.M.); (O.M.V.); (C.S.-R.)
| | - Oscar M. Vidal
- Department of Medicine, Universidad del Norte, Barranquilla 081007, Colombia; (L.C.M.); (O.M.V.); (C.S.-R.)
| | - Ernesto Barceló
- Instituto Colombiano de Neuropedagogía, Barranquilla 080020, Colombia;
| | - Carlos Silvera-Redondo
- Department of Medicine, Universidad del Norte, Barranquilla 081007, Colombia; (L.C.M.); (O.M.V.); (C.S.-R.)
| | - Jorge I. Vélez
- Department of Industrial Engineering, Universidad del Norte, Barranquilla 081007, Colombia;
| | - Pilar Garavito-Galofre
- Department of Medicine, Universidad del Norte, Barranquilla 081007, Colombia; (L.C.M.); (O.M.V.); (C.S.-R.)
| |
Collapse
|
37
|
Gebara N, Correia Y, Wang K, Bussolati B. Angiogenic Properties of Placenta-Derived Extracellular Vesicles in Normal Pregnancy and in Preeclampsia. Int J Mol Sci 2021; 22:5402. [PMID: 34065595 PMCID: PMC8160914 DOI: 10.3390/ijms22105402] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/14/2021] [Accepted: 05/17/2021] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis is one of the main processes that coordinate the biological events leading to a successful pregnancy, and its imbalance characterizes several pregnancy-related diseases, including preeclampsia. Intracellular interactions via extracellular vesicles (EVs) contribute to pregnancy's physiology and pathophysiology, and to the fetal-maternal interaction. The present review outlines the implications of EV-mediated crosstalk in the angiogenic process in healthy pregnancy and its dysregulation in preeclampsia. In particular, the effect of EVs derived from gestational tissues in pro and anti-angiogenic processes in the physiological and pathological setting is described. Moreover, the application of EVs from placental stem cells in the clinical setting is reported.
Collapse
Affiliation(s)
- Natalia Gebara
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10124 Torino, Italy;
| | - Yolanda Correia
- Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham B4 7ET, UK; (Y.C.); (K.W.)
| | - Keqing Wang
- Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham B4 7ET, UK; (Y.C.); (K.W.)
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10124 Torino, Italy;
| |
Collapse
|