1
|
Hassaan NA, Mansour HA. Exosomal therapy is a luxury area for regenerative medicine. Tissue Cell 2024; 91:102570. [PMID: 39383641 DOI: 10.1016/j.tice.2024.102570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/02/2024] [Accepted: 09/17/2024] [Indexed: 10/11/2024]
Abstract
Stem cell-based therapies have made significant advancements in tissue regeneration and medical engineering. However, there are limitations to cell transplantation therapy, such as immune rejection and limited cell viability. These limitations greatly impede the translation of stem cell-based tissue regeneration into clinical practice. In recent years, exosomes, which are packaged vesicles released from cells, have shown promising progress. Specifically, exosomes derived from stem cells have demonstrated remarkable therapeutic benefits. Exosomes are nanoscale extracellular vesicles that act as paracrine mediators. They transfer functional cargos, such as miRNA and mRNA molecules, peptides, proteins, cytokines, and lipids, from MSCs to recipient cells. By participating in intercellular communication events, exosomes contribute to the healing of injured or diseased tissues and organs. Studies have shown that the therapeutic effects of MSCs in various experimental paradigms can be solely attributed to their exosomes. Consequently, MSC-derived exosomes can be modified and utilized to develop a unique cell-free therapeutic approach for treating multiple diseases, including neurological, immunological, heart, and other diseases. This review is divided into several categories, including the current understanding of exosome biogenesis, isolation techniques, and their application as therapeutic tools.
Collapse
Affiliation(s)
- Nahla A Hassaan
- Department of Zoology, Faculty of Science, Al-Azhar University, Cairo, Egypt.
| | - Hanaa A Mansour
- Department of Pharmacology, National Organization for Drug Control and Research (NODCAR), Giza, Egypt
| |
Collapse
|
2
|
Levy E, Reilly JP. Pharmacologic Treatments in Acute Respiratory Failure. Crit Care Clin 2024; 40:275-289. [PMID: 38432696 DOI: 10.1016/j.ccc.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
Acute respiratory failure relies on supportive care using non-invasive and invasive oxygen and ventilatory support. Pharmacologic therapies for the most severe form of respiratory failure, acute respiratory distress syndrome (ARDS), are limited. This review focuses on the most promising therapies for ARDS, targeting different mechanisms that contribute to dysregulated inflammation and resultant hypoxemia. Significant heterogeneity exists within the ARDS population. Treatment requires prompt recognition of ARDS and an understanding of which patients may benefit most from specific pharmacologic interventions. The key to finding effective pharmacotherapies for ARDS may rely on deeper understanding of pathophysiology and bedside identification of ARDS subphenotypes.
Collapse
Affiliation(s)
- Elizabeth Levy
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, 3400 Spruce Street, Philadelphia, PA 19146, USA
| | - John P Reilly
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, 3400 Spruce Street, Philadelphia, PA 19146, USA.
| |
Collapse
|
3
|
Ghonim MA, Boyd DF, Flerlage T, Thomas PG. Pulmonary inflammation and fibroblast immunoregulation: from bench to bedside. J Clin Invest 2023; 133:e170499. [PMID: 37655660 PMCID: PMC10471178 DOI: 10.1172/jci170499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023] Open
Abstract
In recent years, there has been an explosion of interest in how fibroblasts initiate, sustain, and resolve inflammation across disease states. Fibroblasts contain heterogeneous subsets with diverse functionality. The phenotypes of these populations vary depending on their spatial distribution within the tissue and the immunopathologic cues contributing to disease progression. In addition to their roles in structurally supporting organs and remodeling tissue, fibroblasts mediate critical interactions with diverse immune cells. These interactions have important implications for defining mechanisms of disease and identifying potential therapeutic targets. Fibroblasts in the respiratory tract, in particular, determine the severity and outcome of numerous acute and chronic lung diseases, including asthma, chronic obstructive pulmonary disease, acute respiratory distress syndrome, and idiopathic pulmonary fibrosis. Here, we review recent studies defining the spatiotemporal identity of the lung-derived fibroblasts and the mechanisms by which these subsets regulate immune responses to insult exposures and highlight past, current, and future therapeutic targets with relevance to fibroblast biology in the context of acute and chronic human respiratory diseases. This perspective highlights the importance of tissue context in defining fibroblast-immune crosstalk and paves the way for identifying therapeutic approaches to benefit patients with acute and chronic pulmonary disorders.
Collapse
Affiliation(s)
- Mohamed A. Ghonim
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
- Department of Microbiology and Immunology, Faculty of Pharmacy, Al Azhar University, Cairo, Egypt
| | - David F. Boyd
- Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, California, USA
| | - Tim Flerlage
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Paul G. Thomas
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
4
|
Mehta KJ. Iron-Related Genes and Proteins in Mesenchymal Stem Cell Detection and Therapy. Stem Cell Rev Rep 2023; 19:1773-1784. [PMID: 37269528 PMCID: PMC10238768 DOI: 10.1007/s12015-023-10569-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2023] [Indexed: 06/05/2023]
Abstract
Mesenchymal stem cells (MSCs) are located in various tissues of the body. These cells exhibit regenerative and reparative properties, which makes them highly valuable for cell-based therapy. Despite this, majority of MSC-related studies remain to be translated for regular clinical use. This is partly because there are methodical challenges in pre-administration MSC labelling, post-administration detection and tracking of cells, and in retention of maximal therapeutic potential in-vivo. This calls for exploration of alternative or adjunctive approaches that would enable better detection of transplanted MSCs via non-invasive methods and enhance MSC therapeutic potential in-vivo. Interestingly, these attributes have been demonstrated by some iron-related genes and proteins.Accordingly, this unique forward-looking article integrates the apparently distinct fields of iron metabolism and MSC biology, and reviews the utility of iron-related genes and iron-related proteins in facilitating MSC detection and therapy, respectively. Effects of genetic overexpression of the iron-related proteins ferritin, transferrin receptor-1 and MagA in MSCs and their utilisation as reporter genes for improving MSC detection in-vivo are critically evaluated. In addition, the beneficial effects of the iron chelator deferoxamine and the iron-related proteins haem oxygenase-1, lipocalin-2, lactoferrin, bone morphogenetic protein-2 and hepcidin in enhancing MSC therapeutics are highlighted with the consequent intracellular alterations in MSCs. This review aims to inform both regenerative and translational medicine. It can aid in formulating better methodical approaches that will improve, complement, or provide alternatives to the current pre-transplantation MSC labelling procedures, and enhance MSC detection or augment the post-transplantation MSC therapeutic potential.
Collapse
Affiliation(s)
- Kosha J Mehta
- Centre for Education, Faculty of Life Sciences and Medicine, King's College London, London, UK.
| |
Collapse
|
5
|
Taenaka H, Matthay MA. Mechanisms of impaired alveolar fluid clearance. Anat Rec (Hoboken) 2023:10.1002/ar.25166. [PMID: 36688689 PMCID: PMC10564110 DOI: 10.1002/ar.25166] [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: 10/19/2022] [Revised: 12/09/2022] [Accepted: 01/04/2023] [Indexed: 01/24/2023]
Abstract
Impaired alveolar fluid clearance (AFC) is an important cause of alveolar edema fluid accumulation in patients with acute respiratory distress syndrome (ARDS). Alveolar edema leads to insufficient gas exchange and worse clinical outcomes. Thus, it is important to understand the pathophysiology of impaired AFC in order to develop new therapies for ARDS. Over the last few decades, multiple experimental studies have been done to understand the molecular, cellular, and physiological mechanisms that regulate AFC in the normal and the injured lung. This review provides a review of AFC in the normal lung, focuses on the mechanisms of impaired AFC, and then outlines the regulation of AFC. Finally, we summarize ongoing challenges and possible future research that may offer promising therapies for ARDS.
Collapse
Affiliation(s)
- Hiroki Taenaka
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California, USA
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, California, USA
- Department of Anesthesiology and Intensive Care Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Michael A. Matthay
- Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, California, USA
- Department of Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, California, USA
| |
Collapse
|
6
|
Hu Y, Shao J, Shen L, Wang S, Xu K, Mao J, Shen J, Chen W. Protection of adipose-derived mesenchymal stromal cells during acute lung injury requires autophagy maintained by mTOR. Cell Death Dis 2022; 8:481. [PMID: 36470863 PMCID: PMC9722689 DOI: 10.1038/s41420-022-01267-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 09/15/2022] [Accepted: 11/18/2022] [Indexed: 12/10/2022]
Abstract
Previous studies suggest that mesenchymal stem cells may represent a promising cellular therapy for acute lung injury (ALI); however, the underlying relevant molecular mechanisms remain unclear. Adipose-derived mesenchymal stem cells (ADSCs) were isolated and characterized by alizarin red staining, oil red staining, and flow cytometry. Lung injury and inflammatory cell infiltration were determined using the Evans blue method, wet/dry weight ratio, and H&E staining. An ELISA was used to detect the concentrations of IFN-γ, IL-2, and TNF-α. Autophagy was detected with an mRFP-GFP-LC3 dual-fluorescence autophagy indicator system, Western blotting, and electron microscopy. We first demonstrated that ADSCs did alleviate the inflammatory responses and tissue damage in lipopolysaccharide (LPS)-induced ALI. Next, we further demonstrated in vivo that autophagy plays a key role in the maintenance of ADSC therapeutic efficacy. In vitro experiments demonstrated that ADSCs co-cultured with alveolar epithelial cells depend on autophagy for significant anti-inflammatory functions. Moreover, the mammalian target of rapamycin (mTOR) is a key regulator of autophagy. Taken together, our findings demonstrate that the effect of ADSC on ALI, especially on alveolar epithelial cells, is dependent on mTOR-mediated autophagy maintenance. The significance of our study for ALI therapy is discussed with respect to a more complete understanding of the therapeutic strategy paradigm.
Collapse
Affiliation(s)
- Yue Hu
- grid.412465.0Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, 310009 Hangzhou, Zhejiang China
| | - Jing Shao
- Cancer Institute of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, 310012 Hangzhou, Zhejiang China
| | - Lanying Shen
- grid.412465.0Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, 310009 Hangzhou, Zhejiang China
| | - Shengchao Wang
- grid.13402.340000 0004 1759 700XDepartment of Gynecological Oncology, Women’s Hospital, Zhejiang University School of Medicine, 310006 Hangzhou, Zhejiang China
| | - Kaiyan Xu
- grid.412465.0Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, 310009 Hangzhou, Zhejiang China
| | - Jiayan Mao
- Cancer Institute of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, 310012 Hangzhou, Zhejiang China
| | - Jian Shen
- Cancer Institute of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, 310012 Hangzhou, Zhejiang China
| | - Wei Chen
- Cancer Institute of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, 310012 Hangzhou, Zhejiang China
| |
Collapse
|
7
|
Cao C, Zhang L, Liu F, Shen J. Therapeutic Benefits of Mesenchymal Stem Cells in Acute Respiratory Distress Syndrome: Potential Mechanisms and Challenges. J Inflamm Res 2022; 15:5235-5246. [PMID: 36120184 PMCID: PMC9473549 DOI: 10.2147/jir.s372046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/31/2022] [Indexed: 11/23/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) presents as a form of acute respiratory failure resulting from non-cardiogenic pulmonary edema due to excessive alveolocapillary permeability, which may be pulmonary or systemic in origin. In the last 3 years, the coronavirus disease 2019 pandemic has resulted in an increase in ARDS cases and highlighted the challenges associated with this syndrome, as well as the unacceptably high mortality rates and lack of effective treatments. Currently, clinical treatment remains primarily supportive, including mechanical ventilation and drug-based therapy. Mesenchymal stem cell (MSC) therapies are emerging as a promising intervention in patients with ARDS and have promising therapeutic effects and safety. The therapeutic mechanisms include modifying the immune response and assisting with tissue repair. This review provides an overview of the general properties of MSCs and outlines their role in mitigating lung injury and promoting tissue repair in ARDS. Finally, we summarize the current challenges in the study of translational MSC research and identify avenues by which the discipline may progress in the coming years.
Collapse
Affiliation(s)
- Chao Cao
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai, People's Republic of China.,Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai, People's Republic of China.,Center of Emergency and Critical Medicine in Jinshan Hospital of Fudan University, Shanghai, People's Republic of China.,Shanghai Medical College Fudan University, Shanghai, People's Republic of China
| | - Lin Zhang
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai, People's Republic of China.,Center of Emergency and Critical Medicine in Jinshan Hospital of Fudan University, Shanghai, People's Republic of China
| | - Fuli Liu
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai, People's Republic of China.,Center of Emergency and Critical Medicine in Jinshan Hospital of Fudan University, Shanghai, People's Republic of China
| | - Jie Shen
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai, People's Republic of China.,Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai, People's Republic of China.,Center of Emergency and Critical Medicine in Jinshan Hospital of Fudan University, Shanghai, People's Republic of China.,Shanghai Medical College Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
8
|
Rasaei R, Tyagi A, Rasaei S, Lee SJ, Yang SR, Kim KS, Ramakrishna S, Hong SH. Human pluripotent stem cell-derived macrophages and macrophage-derived exosomes: therapeutic potential in pulmonary fibrosis. Stem Cell Res Ther 2022; 13:433. [PMID: 36056418 PMCID: PMC9438152 DOI: 10.1186/s13287-022-03136-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 08/14/2022] [Indexed: 11/10/2022] Open
Abstract
Pulmonary fibrosis (PF) is a fatal chronic disease characterized by accumulation of extracellular matrix and thickening of the alveolar wall, ultimately leading to respiratory failure. PF is thought to be initiated by the dysfunction and aberrant activation of a variety of cell types in the lung. In particular, several studies have demonstrated that macrophages play a pivotal role in the development and progression of PF through secretion of inflammatory cytokines, growth factors, and chemokines, suggesting that they could be an alternative therapeutic source as well as therapeutic target for PF. In this review, we describe the characteristics, functions, and origins of subsets of macrophages involved in PF and summarize current data on the generation and therapeutic application of macrophages derived from pluripotent stem cells for the treatment of fibrotic diseases. Additionally, we discuss the use of macrophage-derived exosomes to repair fibrotic lung tissue.
Collapse
Affiliation(s)
- Roya Rasaei
- Department of Internal Medicine, School of Medicine, Kangwon National University, 1 Kangwondaehakgil, Chuncheon, Gangwon-do, 24431, South Korea
| | - Apoorvi Tyagi
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, South Korea
| | - Shima Rasaei
- Department of Cellular and Molecular Science, Falavarjan Branch, Islamic Azad University, Falavarjan, Iran
| | - Seung-Joon Lee
- Department of Internal Medicine, School of Medicine, Kangwon National University, 1 Kangwondaehakgil, Chuncheon, Gangwon-do, 24431, South Korea
| | - Se-Ran Yang
- Department of Thoracic and Cardiology, School of Medicine, Kangwon National University, Chuncheon, 24341, South Korea
| | - Kye-Seong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, South Korea
| | - Suresh Ramakrishna
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, 04763, South Korea.
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, 1 Kangwondaehakgil, Chuncheon, Gangwon-do, 24431, South Korea.
- Institute of Medical Science, Kangwon National University, Chuncheon, 24341, South Korea.
- KW-Bio Co., Ltd, Wonju, South Korea.
| |
Collapse
|
9
|
Sharma A, Kulkarni R, Sane H, Awad N, Bopardikar A, Joshi A, Baweja S, Joshi M, Vishwanathan C, Gokulchandran N, Badhe P, Khan M, Paranjape A, Kulkarni P, Methal AK. Phase 1 clinical trial for intravenous administration of mesenchymal stem cells derived from umbilical cord and placenta in patients with moderate COVID-19 virus pneumonia: results of stage 1 of the study. AMERICAN JOURNAL OF STEM CELLS 2022; 11:37-55. [PMID: 35873716 PMCID: PMC9301142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/10/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVE Mesenchymal stem cells can serve as a therapeutic option for COVID-19. Their immunomodulatory and anti-inflammatory properties can regulate the exaggerated inflammatory response and promote recovery of lung damage. METHOD Phase-1, single-centre open-label, prospective clinical trial was conducted to evaluate the safety and efficacy of intravenous administration of mesenchymal stem cells derived from umbilical cord and placenta in moderate COVID-19. The study was done in 2 stages with total 20 patients. Herein, the results of stage 1 including first 10 patients receiving 100 million cells on day 1 and 4 with a follow up of 6 months have been discussed. RESULTS No adverse events were recorded immediately after the administration of MSCs or on follow up. There was no deterioration observed in clinical, laboratory and radiological parameters. All symptoms of the study group resolved within 10 days. Levels of inflammatory biomarkers such as NLR, CRP, IL6, ferritin and D-dimer improved in all patients after intervention along with improved oxygenation demonstrated by improvement in the SpO2/FiO2 ratio and PaO2/FiO2 ratio. None of the patients progressed to severe stage. 9 out of 10 patients were discharged within 9 days of their admission. Improvements were noted in chest x-ray and chest CT scan scores at day 7 in most patients. No post-covid fibrosis was observed on chest CT 28 days after intervention and Chest X ray after 6 months of the intervention. CONCLUSION Administration of 100 million mesenchymal stem cells in combination with standard treatment was found to be safe and resulted in prevention of the cytokine storm, halting of the disease progression and acceleration of recovery in moderate COVID-19. This clinical trial has been registered with the Clinical Trial Registry- India (CTRI) as CTRI/2020/08/027043. http://www.ctri.nic.in/Clinicaltrials/pmaindet2.php?trialid=43175.
Collapse
Affiliation(s)
- Alok Sharma
- Department of Neurosurgery, LTMG Hospital and LTM Medical CollegeMumbai, Maharashtra, India
- Department of Medical Services and Clinical Research, NeuroGen Brain & Spine InstituteNavi Mumbai, Maharashtra, India
| | | | - Hemangi Sane
- Department of Research & Development, NeuroGen Brain & Spine InstituteNavi Mumbai, Maharashtra, India
| | - Nilkanth Awad
- Department of Pulmonary Medicine, LTMG Hospital and LTM Medical CollegeSion, Mumbai, Maharashtra, India
| | | | - Anagha Joshi
- Department of Radiology, LTMG Hospital and LTM Medical CollegeMumbai, Maharashtra, India
| | - Sujata Baweja
- Department of Microbiology, LTMG Hospital and LTM Medical CollegeMumbai, Maharashtra, India
| | - Mohan Joshi
- Dean, LTMG Hospital and LTM Medical CollegeMumbai, Maharashtra, India
| | | | - Nandini Gokulchandran
- Department of Medical Services and Clinical Research, NeuroGen Brain & Spine InstituteNavi Mumbai, Maharashtra, India
| | - Prerna Badhe
- Department of Regenerative Laboratory, NeuroGen Brain and Spine InstituteSeawoods, Navi Maharashtra, India
| | - Mazhar Khan
- Department of Neurosurgery, LTMG Hospital and LTM Medical CollegeMumbai, Maharashtra, India
| | - Amruta Paranjape
- Department of Research & Development, NeuroGen Brain & Spine InstituteNavi Mumbai, Maharashtra, India
| | - Pooja Kulkarni
- Department of Research & Development, NeuroGen Brain & Spine InstituteNavi Mumbai, Maharashtra, India
| | - Arjun K Methal
- Department of Research & Development, NeuroGen Brain & Spine InstituteNavi Mumbai, Maharashtra, India
| |
Collapse
|
10
|
Ngai HW, Kim DH, Hammad M, Gutova M, Aboody K, Cox CD. Stem Cell-based therapies for COVID-19-related acute respiratory distress syndrome. J Cell Mol Med 2022; 26:2483-2504. [PMID: 35426198 PMCID: PMC9077311 DOI: 10.1111/jcmm.17265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/24/2022] [Accepted: 02/28/2022] [Indexed: 11/30/2022] Open
Abstract
As the number of confirmed cases and resulting death toll of the COVID-19 pandemic continue to increase around the globe - especially with the emergence of new mutations of the SARS-CoV-2 virus in addition to the known alpha, beta, gamma, delta and omicron variants - tremendous efforts continue to be dedicated to the development of interventive therapeutics to mitigate infective symptoms or post-viral sequelae in individuals for which vaccines are not accessible, viable or effective in the prevention of illness. Many of these investigations aim to target the associated acute respiratory distress syndrome, or ARDS, which induces damage to lung epithelia and other physiologic systems and is associated with progression in severe cases. Recently, stem cell-based therapies have demonstrated preliminary efficacy against ARDS based on a number of preclinical and preliminary human safety studies, and based on promising outcomes are now being evaluated in phase II clinical trials for ARDS. A number of candidate stem cell therapies have been found to exhibit low immunogenicity, coupled with inherent tropism to injury sites. In recent studies, these have demonstrated the ability to modulate suppression of pro-inflammatory cytokine signals such as those characterizing COVID-19-associated ARDS. Present translational studies are aiming to optimize the safety, efficacy and delivery to fully validate stem cell-based strategies targeting COVID-19 associated ARDS for viable clinical application.
Collapse
Affiliation(s)
- Hoi Wa Ngai
- Department of Stem Cell Biology and Regenerative MedicineCity of Hope Beckman Research InstituteDuarteCaliforniaUSA
| | - Dae Hong Kim
- Department of Stem Cell Biology and Regenerative MedicineCity of Hope Beckman Research InstituteDuarteCaliforniaUSA
| | - Mohamed Hammad
- Department of Stem Cell Biology and Regenerative MedicineCity of Hope Beckman Research InstituteDuarteCaliforniaUSA
| | - Margarita Gutova
- Department of Stem Cell Biology and Regenerative MedicineCity of Hope Beckman Research InstituteDuarteCaliforniaUSA
| | - Karen Aboody
- Department of Stem Cell Biology and Regenerative MedicineCity of Hope Beckman Research InstituteDuarteCaliforniaUSA
| | | |
Collapse
|
11
|
N V Lakshmi Kavya A, Subramanian S, Ramakrishna S. Therapeutic applications of exosomes in various diseases: A review. BIOMATERIALS ADVANCES 2022; 134:112579. [PMID: 35525729 DOI: 10.1016/j.msec.2021.112579] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 11/10/2021] [Accepted: 11/25/2021] [Indexed: 06/14/2023]
Abstract
Exosomes (30-150 nm in diameter) a subset of extracellular vesicles, secreted by mostly all cells, have been gaining enormous recognition from the last decade. In recent times, several studies have included exosomes to design novel therapeutic applications along with their contribution to diagnostic evaluations and pathophysiological processes. Based on cell origin, they show diverse functions and characteristics. This article is classified into several sections that include exosomes biogenesis, isolation methods, and application as therapeutic tools, commercialized exosome products, clinical trials, benefits, and challenges faced in the progress of exosome-dependent therapeutics. This work aims to give a thorough review of the numerous studies where exosomes act as therapeutic tools in the treatment of various disorders including heart, kidney, liver, and lung illnesses. The clinical trials involving exosomes, their advantages, and hazards, and difficulties involved during storage and large-scale production, applications of nanotechnology in exosome research while applying for therapeutic applications, and future directions are summarized.
Collapse
Affiliation(s)
| | - Sundarrajan Subramanian
- Center for Nanofibers and Nanotechnology Lab, Mechanical Engineering, National University of Singapore, Blk E3 05-12, 2 Engineering Drive 3, Singapore 117581, Singapore.
| | - Seeram Ramakrishna
- Center for Nanofibers and Nanotechnology Lab, Mechanical Engineering, National University of Singapore, Blk E3 05-12, 2 Engineering Drive 3, Singapore 117581, Singapore.
| |
Collapse
|
12
|
Sanie-Jahromi F, NejatyJahromy Y, Jahromi RR. A Review on the Role of Stem Cells against SARS-CoV-2 in Children and Pregnant Women. Int J Mol Sci 2021; 22:11787. [PMID: 34769218 PMCID: PMC8584228 DOI: 10.3390/ijms222111787] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/10/2021] [Accepted: 10/28/2021] [Indexed: 12/18/2022] Open
Abstract
Since the COVID-19 outbreak was acknowledged by the WHO on 30 January 2020, much research has been conducted to unveil various features of the responsible SARS-CoV-2 virus. Different rates of contagion in adults, children, and pregnant women may guide us to understand the underlying infection conditions of COVID-19. In this study, we first provide a review of recent reports of COVID-19 clinical outcomes in children and pregnant women. We then suggest a mechanism that explains the curious case of COVID-19 in children/pregnant women. The unique stem cell molecular signature, as well as the very low expression of angiotensin-converting enzyme 2 and the lower ACE/ACE2 ratio in stem cells of children/pregnant women compared to adults might be the cause of milder symptoms of COVID-19 in them. This study provides the main molecular keys on how stem cells can function properly and exert their immunomodulatory and regenerative effects in COVID-19-infected children/pregnant women, while failing to replicate their role in adults. This can lay the groundwork for both predicting the pattern of spread and severity of the symptoms in a population and designing novel stem cell-based treatment and prevention strategies for COVID-19.
Collapse
Affiliation(s)
- Fatemeh Sanie-Jahromi
- Poostchi Ophthalmology Research Center, Shiraz University of Medical Sciences, Shiraz 7134997446, Iran;
| | - Yaser NejatyJahromy
- Institut für Physikalische und Theoretische Chemie, Rheinische Friedrich-Wilhelms-Universität Bonn, 53012 Bonn, Germany
| | - Rahim Raoofi Jahromi
- Department of Infectious Disease, Peymanieh Hospital, Jahrom University of Medical Science, Jahrom 7414846199, Iran
| |
Collapse
|
13
|
Abstract
Acute respiratory distress syndrome is a heterogenous syndrome with many etiologies for which there are no definitive pharmacologic treatments, despite decades of research. We explore some adjunctive pharmacologic therapies, including neuromuscular blockade, corticosteroids, and inhaled pulmonary vasodilators. Additionally, we explore some investigative therapies, including Vitamin C, beta-agonists, statins, mesenchymal stromal cells, and granulocyte-macrophage colony stimulating factor. We do discuss the potential role of steroids in acute respiratory distress syndrome with severe acute respiratory syndrome coronavirus 2 as a trigger. The standard of care, however, remains supportive care.
Collapse
Affiliation(s)
- Nida Qadir
- David Geffen School of Medicine at UCLA, 10833 Le Conte Avenue, Room 43-229 CHS, Los Angeles, CA 90095, USA.
| | - Steven Y Chang
- David Geffen School of Medicine at UCLA, 10833 Le Conte Avenue, Room 43-229 CHS, Los Angeles, CA 90095, USA
| |
Collapse
|
14
|
Afarid M, Sanie-Jahromi F. Mesenchymal Stem Cells and COVID-19: Cure, Prevention, and Vaccination. Stem Cells Int 2021; 2021:6666370. [PMID: 34035820 PMCID: PMC8103964 DOI: 10.1155/2021/6666370] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/26/2021] [Accepted: 04/21/2021] [Indexed: 02/07/2023] Open
Abstract
COVID-19 disease has been a global health problem since late 2019. There are many concerns about the rapid spread of this disease, and yet, there is no approved treatment for COVID-19. Several biological interventions have been under study recently to investigate efficient treatment for this viral disease. Besides, many efforts have been made to find a safe way to prevent and vaccinate people against COVID-19 disease. In severe cases, patients suffer from acute respiratory distress syndrome usually associated with an increased level of inflammatory cytokines, called a cytokine storm. It seems that reequilibrating the hyperinflammatory response of the host immune system and regeneration of damaged cells could be the main way to manage the disease. Mesenchymal stem cells (MSCs) have been recently under investigation in this regard, and the achieved clinical outcomes show promising evidence for stem cell-based therapy of COVID-19. MSCs are known for their potential for immunomodulation, defense against virus infection, and tissue regeneration. MSCs are a newly emerged platform for designing vaccines and show promising evidence in this area. In the present study, we provided a thorough research study on the most recent clinical studies based on stem cells in the treatment of COVID-19 while introducing stem cell exclusivities for use as an immune disorder or lung cell therapy and its potential application for protection and vaccination against COVID-19.
Collapse
Affiliation(s)
- Mehrdad Afarid
- Poostchi Ophthalmology Research Center, Department of Ophthalmology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Sanie-Jahromi
- Poostchi Ophthalmology Research Center, Department of Ophthalmology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
15
|
Alvites RD, Branquinho MV, Sousa AC, Lopes B, Sousa P, Mendonça C, Atayde LM, Maurício AC. Small Ruminants and Its Use in Regenerative Medicine: Recent Works and Future Perspectives. BIOLOGY 2021; 10:biology10030249. [PMID: 33810087 PMCID: PMC8004958 DOI: 10.3390/biology10030249] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/16/2021] [Accepted: 03/18/2021] [Indexed: 12/16/2022]
Abstract
Simple Summary Small ruminants such as sheep and goats have been increasingly used as animal models due to their dimensions, physiology and anatomy identical to those of humans. Their low costs, ease of accommodation, great longevity and easy handling make them advantageous animals to be used in a wide range of research work. Although there is already a lot of scientific literature describing these species, their use still lacks some standardization. The purpose of this review is to summarize the general principles related to the use of small ruminants as animal models for scientific research. Abstract Medical and translational scientific research requires the use of animal models as an initial approach to the study of new therapies and treatments, but when the objective is an exploration of translational potentialities, classical models fail to adequately mimic problems in humans. Among the larger animal models that have been explored more intensely in recent decades, small ruminants, namely sheep and goats, have emerged as excellent options. The main advantages associated to the use of these animals in research works are related to their anatomy and dimensions, larger than conventional laboratory animals, but very similar to those of humans in most physiological systems, in addition to their low maintenance and feeding costs, tendency to be docile, long life expectancies and few ethical complications raised in society. The most obvious disadvantages are the significant differences in some systems such as the gastrointestinal, and the reduced amount of data that limits the comparison between works and the validation of the characterization essays. Despite everything, recently these species have been increasingly used as animal models for diseases in different systems, and the results obtained open doors for their more frequent and advantageous use in the future. The purpose of this review is to summarize the general principles related to the use of small ruminants as animal models, with a focus on regenerative medicine, to group the most relevant works and results published recently and to highlight the potentials for the near future in medical research.
Collapse
Affiliation(s)
- Rui Damásio Alvites
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (C.M.); (L.M.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal
| | - Mariana Vieira Branquinho
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (C.M.); (L.M.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal
| | - Ana Catarina Sousa
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (C.M.); (L.M.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal
| | - Bruna Lopes
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (C.M.); (L.M.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal
| | - Patrícia Sousa
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (C.M.); (L.M.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal
| | - Carla Mendonça
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (C.M.); (L.M.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal
| | - Luís Miguel Atayde
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (C.M.); (L.M.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal
| | - Ana Colette Maurício
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente (ICETA) da Universidade do Porto, Praça Gomes Teixeira, 4051-401 Porto, Portugal; (R.D.A.); (M.V.B.); (A.C.S.); (B.L.); (P.S.); (C.M.); (L.M.A.)
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal
- Correspondence: ; Tel.: +351-919-071-286 or +351-220-428-000
| |
Collapse
|
16
|
Majolo F, da Silva GL, Vieira L, Timmers LFSM, Laufer S, Goettert MI. Review of Trials Currently Testing Stem Cells for Treatment of Respiratory Diseases: Facts Known to Date and Possible Applications to COVID-19. Stem Cell Rev Rep 2021; 17:44-55. [PMID: 32827081 PMCID: PMC7442550 DOI: 10.1007/s12015-020-10033-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Therapeutic clinical and preclinical studies using cultured cells are on the rise, especially now that the World Health Organization (WHO) declared coronavirus disease 2019 (COVID-19) a "public health emergency of international concern", in January, 2020. Thus, this study aims to review the outcomes of ongoing clinical studies on stem cells in Severe Acute Respiratory Syndrome (SARS), Acute Respiratory Distress Syndrome (ARDS), and Middle East Respiratory Syndrome (MERS). The results will be associated with possible applications to COVID-19. Only three clinical trials related to stem cells are considered complete, whereby two are in Phase 1 and one is in Phase 2. Basically, the ongoing studies on coronavirus are using mesenchymal stem cells (MSCs) derived from bone marrow or the umbilical cord to demonstrate their feasibility, safety, and tolerability. The studies not related to coronavirus are all in ARDS conditions; four of them are in Phase 1 and three in Phase 2. With the COVID-19 boom, many clinical trials are being carried out using different sources with an emphasis on MSC-based therapy used to inhibit inflammation. One of the biggest challenges in the current treatment of COVID-19 is the cytokine storm, however MSCs can prevent or mitigate this cytokine storm through their immunomodulatory capacity. We look forward to the results of the ongoing clinical trials to find a treatment for the disease. Researchers around the world are joining forces to help fight COVID-19. Stem cells used in the current clinical studies are a new therapeutic promise for COVID-19 where pharmacological treatments seem insufficient.Graphical Abstract.
Collapse
Affiliation(s)
- Fernanda Majolo
- Post-graduate Program in Biotechnology, Universidade do Vale do Taquari - Univates, Av. Avelino Talini, 171, 95914-014, Lajeado, Rio Grande do Sul, Brazil
| | - Guilherme Liberato da Silva
- Medical Sciences Center, Universidade do Vale do Taquari - Univates, Lajeado, Rio Grande do Sul, 95914-014, Brazil
| | - Lucas Vieira
- Medical Sciences Center, Universidade do Vale do Taquari - Univates, Lajeado, Rio Grande do Sul, 95914-014, Brazil
| | - Luís Fernando Saraiva Macedo Timmers
- Post-graduate Program in Biotechnology, Universidade do Vale do Taquari - Univates, Av. Avelino Talini, 171, 95914-014, Lajeado, Rio Grande do Sul, Brazil
| | - Stefan Laufer
- Medicinal Chemistry, University of Tuebingen, D-72076, Tubingen, Germany
| | - Márcia Inês Goettert
- Post-graduate Program in Biotechnology, Universidade do Vale do Taquari - Univates, Av. Avelino Talini, 171, 95914-014, Lajeado, Rio Grande do Sul, Brazil.
| |
Collapse
|
17
|
Jamshidi E, Babajani A, Soltani P, Niknejad H. Proposed Mechanisms of Targeting COVID-19 by Delivering Mesenchymal Stem Cells and Their Exosomes to Damaged Organs. Stem Cell Rev Rep 2021; 17:176-192. [PMID: 33432484 PMCID: PMC7799400 DOI: 10.1007/s12015-020-10109-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2020] [Indexed: 12/13/2022]
Abstract
With the outbreak of coronavirus disease (COVID-19) caused by novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the world has been facing an unprecedented challenge. Considering the lack of appropriate therapy for COVID-19, it is crucial to develop effective treatments instead of supportive approaches. Mesenchymal stem cells (MSCs) as multipotent stromal cells have been shown to possess treating potency through inhibiting or modulating the pathological events in COVID-19. MSCs and their exosomes participate in immunomodulation by controlling cell-mediated immunity and cytokine release. Furthermore, they repair the renin-angiotensin-aldosterone system (RAAS) malfunction, increase alveolar fluid clearance, and reduce the chance of hypercoagulation. Besides the lung, which is the primary target of SARS-CoV-2, the heart, kidney, nervous system, and gastrointestinal tract are also affected by COVID-19. Thus, the efficacy of targeting these organs via different delivery routes of MSCs and their exosomes should be evaluated to ensure safe and effective MSCs administration in COVID-19. This review focuses on the proposed therapeutic mechanisms and delivery routes of MSCs and their exosomes to the damaged organs. It also discusses the possible application of primed and genetically modified MSCs as a promising drug delivery system in COVID-19. Moreover, the recent advances in the clinical trials of MSCs and MSCs-derived exosomes as one of the promising therapeutic approaches in COVID-19 have been reviewed.
Collapse
Affiliation(s)
- Elham Jamshidi
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirhesam Babajani
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Pegah Soltani
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hassan Niknejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Sadeghi S, Soudi S, Shafiee A, Hashemi SM. Mesenchymal stem cell therapies for COVID-19: Current status and mechanism of action. Life Sci 2020; 262:118493. [PMID: 32979360 PMCID: PMC7510562 DOI: 10.1016/j.lfs.2020.118493] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/19/2020] [Accepted: 09/20/2020] [Indexed: 12/12/2022]
Abstract
The outbreak of COVID-19 in December 2019, has become an urgent and serious public health emergency. At present, there is no effective treatment or vaccine for COVID-19. Therefore, there is a crucial unmet need to develop a safe and effective treatment for COVID-19 patients. Mesenchymal stem cells (MSCs) are widely used in basic science and in a variety of clinical trials. MSCs are able to engraft to the damaged tissues after transplantation and promote tissue regeneration, besides MSCs able to secrete immunomodulatory factors that suppress the cytokine storms. Moreover, the contribution of MSCs to prevent cell death and inhibit tissue fibrosis is well established. In the current review article, the potential mechanisms by which MSCs contribute to the treatment of COVID-19 patients are highlighted. Also, current trials that evaluated the potential of MSC-based treatments for COVID-19 are briefly reviewed.
Collapse
Affiliation(s)
- Somaye Sadeghi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Soudi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Abbas Shafiee
- UQ Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, Queensland, Australia.
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
19
|
Kassem DH, Kamal MM. Mesenchymal Stem Cells and Their Extracellular Vesicles: A Potential Game Changer for the COVID-19 Crisis. Front Cell Dev Biol 2020; 8:587866. [PMID: 33102489 PMCID: PMC7554315 DOI: 10.3389/fcell.2020.587866] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 09/09/2020] [Indexed: 12/15/2022] Open
Abstract
Corona virus disease 2019 (COVID-19) is a global public health crisis. The high infectivity of the disease even from non-symptomatic infected patients, together with the lack of a definitive cure or preventive measures are all responsible for disease outbreak. The severity of COVID-19 seems to be mostly dependent on the patients' own immune response. The over-activation of the immune system in an attempt to kill the virus, can cause a "cytokine storm" which in turn can induce acute respiratory distress syndrome (ARDS), as well as multi-organ damage, and ultimately may lead to death. Thus, harnessing the immunomodulatory properties of mesenchymal stem cells (MSCs) to ameliorate that cytokine-storm can indeed provide a golden key for the treatment of COVID-19 patients, especially severe cases. In fact, MSCs transplantation can improve the overall outcome of COVID-19 patients via multiple mechanisms; first through their immunomodulatory effects which will help to regulate the infected patient inflammatory response, second via promoting tissue-repair and regeneration, and third through their antifibrotic effects. All these mechanisms will interplay and intervene together to enhance lung-repair and protect various organs from any damage resulting from exaggerated immune-response. A therapeutic modality which provides all these mechanisms undoubtedly hold a strong potential to help COVID-19 patients even those with the worst condition to hopefully survive and recover.
Collapse
Affiliation(s)
- Dina H. Kassem
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mohamed M. Kamal
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, The British University in Egypt (BUE), Cairo, Egypt
- The Centre for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt (BUE), Cairo, Egypt
| |
Collapse
|
20
|
Muraca M, Pessina A, Pozzobon M, Dominici M, Galderisi U, Lazzari L, Parolini O, Lucarelli E, Perilongo G, Baraldi E. Mesenchymal stromal cells and their secreted extracellular vesicles as therapeutic tools for COVID-19 pneumonia? J Control Release 2020; 325:135-140. [PMID: 32622963 PMCID: PMC7332437 DOI: 10.1016/j.jconrel.2020.06.036] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/12/2020] [Accepted: 06/29/2020] [Indexed: 12/13/2022]
Abstract
The COVID-19 epidemic represents an unprecedented global health emergency, further aggravated by the lack of effective therapies. For this reason, several clinical trials are testing different off-label drugs, already approved for other pathologies. Mesenchymal stem/stromal cells (MSCs) have been tested during the last two decades for the treatment of various pathologic conditions, including acute and chronic lung diseases, both in animal models and in patients. In particular, promising results have been obtained in the experimental therapy of acute respiratory distress syndrome, which represents the most threatening complication of COVID-19 infection. Furthermore, more recently, great interest has been devoted to the possible clinical applications of extracellular vesicles secreted by MSCs, nanoparticles that convey much of the biological effects and of the therapeutic efficacy of their cells of origin. This review summarizes the experimental evidence underlying the possible use of MSCs and of MSC-EVs in severe COVID-19 infection and underlines the need to evaluate the possible efficacy of these therapeutic approaches through controlled studies under the supervision of the Regulatory Authorities.
Collapse
Affiliation(s)
- Maurizio Muraca
- Department of Women's and Children's Health, University of Padova, Italy
| | - Augusto Pessina
- CRC StaMeTec, Department of Biomedical, Surgical and Dental Sciences, University of MilanVia Pascal 3620133 Milano - Italy.
| | - Michela Pozzobon
- Department of Women's and Children's Health, University of Padova, Italy
| | - Massimo Dominici
- Laboratory of Cellular Therapy, University Hospital of Modena and Reggio Emilia, Modena, Italy
| | - Umberto Galderisi
- Department of Experimental Medicine, Luigi Vanvitelli University, Naples, Italy
| | - Lorenza Lazzari
- Laboratory of Regenerative Medicine - Cell Factory, Department of Trasfusion Medicine and Hematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Ornella Parolini
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, and Fondazione Policlinico Universitario A Gemelli IRCCS, Rome, Italy
| | - Enrico Lucarelli
- Osteoarticolar Regeneration Laboratory, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Giorgio Perilongo
- Department of Women's and Children's Health, University of Padova, Italy
| | - Eugenio Baraldi
- Department of Women's and Children's Health, University of Padova, Italy
| |
Collapse
|
21
|
El-Metwaly S, El-Senduny FF, El-Demerdash RS, Abdel-Aziz AF. Mesenchymal stem cells alleviate hydrochloric acid-induced lung injury through suppression of inflammation, oxidative stress and apoptosis in comparison to moxifloxacin and sildenafil. Heliyon 2019; 5:e02710. [PMID: 31872097 PMCID: PMC6909079 DOI: 10.1016/j.heliyon.2019.e02710] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 07/21/2019] [Accepted: 10/18/2019] [Indexed: 12/12/2022] Open
Abstract
Introduction Acute lung injury (ALI) is a severe life-threatening disease causing uncontrolled pulmonary inflammation and oxidative damage. There are still no effective therapies for this disease. The aim of this study was to evaluate the protective role of mesenchymal stem cells, moxifloxacin, sildenafil or a combination of moxifloxacin and sildenafil against hydrochloric Acid (HCl) - induced ALI. Methods HCl or saline was injected intra-tracheally and after 2 h, moxifloxacin, sildenafil, moxifloxacin + sildenafil or mesenchymal stem cells were injected. After 7 days, rats were sacrificed for evaluation of the blood chemistry and inflammation via determination of the level of oxidative stress markers, apoptosis and the histopathological alterations by H&E. Results In HCl-injected rats, there were a significant increase in total white blood cells (WBCs), lymphocytes, malondialdehyde (MDA) and caspase-3 gene expression. Also, there were a significant decrease in superoxide dismutase (SOD), catalase (CAT), reduced glutathione (GSH) and Hemeoxygenase-1 (HO-1) gene expression in lung tissue. On the other hand, treatment of lung injured rats with mesenchymal stem cell, moxifloxacin, sildenafil or a combination of moxifloxacin and sildenafil showed a significant decrease in WBCs and lymphocytes and ameliorated the histopathological changes. MDA level in lung tissue was only significantly lowered in rats treated with moxifloxacin alone or in combination with sildenafil or MSCs. GSH was just increased in rats treated with moxifloxacin, sildenafil or with MSCs. Antioxidant parameters and gene expression of HO-1 and caspase-3 were significantly modulated in rats treated with MSCs. Conclusion MSCs ameliorated the toxic effects of HCl through their ability to decrease inflammation, oxidative stress, and apoptosis in acute lung injury.
Collapse
Affiliation(s)
- Shimaa El-Metwaly
- Chemistry Department, Faculty of Science, Mansoura University, Egypt
| | | | | | - A F Abdel-Aziz
- Chemistry Department, Faculty of Science, Mansoura University, Egypt
| |
Collapse
|
22
|
Exosomal miRNAs in Lung Diseases: From Biologic Function to Therapeutic Targets. J Clin Med 2019; 8:jcm8091345. [PMID: 31470655 PMCID: PMC6781233 DOI: 10.3390/jcm8091345] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 08/26/2019] [Accepted: 08/27/2019] [Indexed: 12/12/2022] Open
Abstract
Increasing evidence suggests the potential role of extracellular vesicles (EVs) in many lung diseases. According to their subcellular origin, secretion mechanism, and size, EVs are currently classified into three subpopulations: exosomes, microvesicles, and apoptotic bodies. Exosomes are released in most biofluids, including airway fluids, and play a key role in intercellular communication via the delivery of their cargo (e.g., microRNAs (miRNAs)) to target cell. In a physiological context, lung exosomes present protective effects against stress signals which allow them to participate in the maintenance of lung homeostasis. The presence of air pollution alters the composition of lung exosomes (dysregulation of exosomal miRNAs) and their homeostatic property. Indeed, besides their potential as diagnostic biomarkers for lung diseases, lung exosomes are functional units capable of dysregulating numerous pathophysiological processes (including inflammation or fibrosis), resulting in the promotion of lung disease progression. Here, we review recent studies on the known and potential role of lung exosomes/exosomal miRNAs, in the maintaining of lung homeostasis on one hand, and in promoting lung disease progression on the other. We will also discuss using exosomes as prognostic/diagnostic biomarkers as well as therapeutic tools for lung diseases.
Collapse
|
23
|
Cardenes N, Aranda-Valderrama P, Carney JP, Sellares Torres J, Alvarez D, Kocyildirim E, Wolfram Smith JA, Ting AE, Lagazzi L, Yu Z, Mason S, Santos E, Lopresti BJ, Rojas M. Cell therapy for ARDS: efficacy of endobronchial versus intravenous administration and biodistribution of MAPCs in a large animal model. BMJ Open Respir Res 2019; 6:e000308. [PMID: 30713713 PMCID: PMC6339992 DOI: 10.1136/bmjresp-2018-000308] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 11/14/2018] [Accepted: 11/14/2018] [Indexed: 12/14/2022] Open
Abstract
Introduction Bone marrow-derived multipotent adult progenitor cells (MAPCs) are adult allogeneic adherent stem cells currently investigated clinically for use in acute respiratory distress syndrome (ARDS). To date, there is no agreement on which is the best method for stem cells delivery in ARDS. Here, we compared the efficacy of two different methods of administration and biodistribution of MAPC for the treatment of ARDS in a sheep model. Methods MAPC were labelled with [18F] fluoro-29-deoxy-D-glucose and delivered by endobronchial (EB) or intravenous route 1 hour after lipopolysaccharide infusion in sheep mechanically ventilated. PET/CT images were acquired to determine the biodistribution and retention of the cells at 1 and 5 hours of administration. Results The distribution and retention of the MAPC was dependent on the method of cell administration. By EB route, PET images showed that MAPC remained at the site of administration and no changes were observed after 5 hours, whereas with intravenous route, the cells had broad biodistribution to different organs, being the lung the main organ of retention at 1 and 5 hours. MAPC demonstrated an equal effect on arterial oxygenation recovery by either route of administration. Conclusion The EB or intravenous routes of administration of MAPC are both effective for the treatment of ARDS in an acute sheep model, and the effect of MAPC therapy is not dependent of parenchymal integration or systemic biodistribution.
Collapse
Affiliation(s)
- Nayra Cardenes
- The Dorothy P. and Richard P. Simmons Center for Interstitial Lung Diseases, Pittsburgh, Pennsylvania, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Paola Aranda-Valderrama
- The Dorothy P. and Richard P. Simmons Center for Interstitial Lung Diseases, Pittsburgh, Pennsylvania, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Jonathan P Carney
- Department of Radiology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Jacobo Sellares Torres
- The Dorothy P. and Richard P. Simmons Center for Interstitial Lung Diseases, Pittsburgh, Pennsylvania, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- Interstitial Lung Disease Program, Servei de Pneumología, Institut clinic respiratori, Hospital Clínic, Barcelona, Spain
| | - Diana Alvarez
- The Dorothy P. and Richard P. Simmons Center for Interstitial Lung Diseases, Pittsburgh, Pennsylvania, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Ergin Kocyildirim
- Department of Cardiothoracic Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Antony E Ting
- Cardiopulmonary Program at Athersys, Inc, Cleveland, Ohio, USA
| | - Luigi Lagazzi
- Department of Cardiothoracic Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Zheming Yu
- Department of Radiology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Scott Mason
- Department of Radiology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Ernesto Santos
- Department of Radiology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Brian J Lopresti
- Department of Radiology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Mauricio Rojas
- The Dorothy P. and Richard P. Simmons Center for Interstitial Lung Diseases, Pittsburgh, Pennsylvania, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
24
|
Chen XX, Tang L, Han ZH, Wang WJ, Meng JG. Coculture with bone marrow‑derived mesenchymal stem cells attenuates inflammation and apoptosis in lipopolysaccharide‑stimulated alveolar epithelial cells via enhanced secretion of keratinocyte growth factor and angiopoietin‑1 modulating the Toll‑like receptor‑4 signal pathway. Mol Med Rep 2019; 19:1891-1902. [PMID: 30628710 DOI: 10.3892/mmr.2019.9836] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 12/11/2018] [Indexed: 11/06/2022] Open
Abstract
Acute lung injury (ALI) is a common, costly and potentially lethal disease with characteristics of alveolar‑capillary membrane disruption, pulmonary edema and impaired gas exchange due to increased apoptosis and pulmonary inflammation. There is no effective and specific therapy for ALI; however, mesenchymal stem cells (MSCs) have been demonstrated to be a potential option. Lipopolysaccharide (LPS) is a highly proinflammatory molecule that is used to mimic an in vivo inflammatory and damaged state in vitro. The present study investigated the effect of bone marrow‑derived MSCs on an LPS‑induced alveolar epithelial cell (A549 cell line) injury and its underlying mechanisms by a Transwell system. It was identified that a high LPS concentration caused a decrease in cell viability, increases in apoptosis, inflammatory cytokine release and NF‑κB activity, disruption of the caspase‑3/Bcl‑2 ratio, upregulation of Toll‑like receptor 4 (TLR4), myeloid differentiation factor 88 (MyD88) and toll‑interleukin‑1 receptor domain‑containing adaptor inducing interferon (TRIF) expression, and facilitation of TLR4/MyD88 and TLR4/TRIF complex formation in A549 cells. Coculture with MSCs attenuated all of these activities induced by LPS in A549 cells. In addition, an increased level of keratinocyte growth factor (KGF) and angiopoietin‑1 (ANGPT1) secretion from MSCs was observed under inflammatory stimulation. KGF and/or ANGPT1 neutralizing antibodies diminished the beneficial effect of MSC conditioned medium. These data suggest that MSCs alleviate inflammatory damage and cellular apoptosis induced by LPS in A549 cells by modulating TLR4 signals. These changes may be partly associated with an increased secretion of KGF and ANGPT1 from MSCs under inflammatory conditions. These data provide the basis for development of MSC‑based therapies for ALI.
Collapse
Affiliation(s)
- Xu-Xin Chen
- Department of Respiratory Medicine, Navy General Hospital of The People's Liberation Army, Beijing 100037, P.R. China
| | - Lu Tang
- Department of Neurology, The First Hospital of Changsha, Changsha, Hunan 430100, P.R. China
| | - Zhi-Hai Han
- Department of Respiratory Medicine, Navy General Hospital of The People's Liberation Army, Beijing 100037, P.R. China
| | - Wen-Jing Wang
- Cardiothoracic Surgery Intensive Care Unit, Anqing Municipal Hospital, Anqing, Anhui 246003, P.R. China
| | - Ji-Guang Meng
- Department of Respiratory Medicine, Navy General Hospital of The People's Liberation Army, Beijing 100037, P.R. China
| |
Collapse
|
25
|
Mokhber Dezfouli MR, Jabbari Fakhr M, Sadeghian Chaleshtori S, Dehghan MM, Vajhi A, Mokhtari R. Intrapulmonary autologous transplant of bone marrow-derived mesenchymal stromal cells improves lipopolysaccharide-induced acute respiratory distress syndrome in rabbit. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2018; 22:353. [PMID: 30572913 PMCID: PMC6302408 DOI: 10.1186/s13054-018-2272-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 11/21/2018] [Indexed: 02/06/2023]
Abstract
Background Lung diseases such as acute respiratory distress syndrome (ARDS) have a high incidence worldwide. The current drug therapies for ARDS have supportive effects, making them inefficient. New methods such as stromal cell therapy are needed for this problem. Methods This research was performed with ten New Zealand rabbits in two groups. Bone marrow aspiration was performed on the treated group, and mesenchymal stem cells were isolated and cultured. The experimental model of ARDS was induced using LPS from Escherichia coli strain O55:B5. Then, 1010 bone marrow mesenchymal stem cells (BM-MSCs) were autologously transplanted intrapulmonary in the treatment group, and 1–2 ml of PBS in the control group. The clinical signs, computed tomographic (CT) scans, echocardiography, blood gas analysis, complete blood count, and cytokine levels were measured before and at 3, 6, 12, 24, 48, 72, and 168 h after BM-MSC transplant. Finally, the rabbits were killed, and histopathological examination was performed. Results The results showed that BM-MSCs decreased the severity of clinical symptoms, the number of white blood cells and heterophils in the blood, the total cell count, and number of heterophils and macrophages in bronchoalveolar lavage, and balanced the values of arterial blood gases (increase in partial pressure of oxygen and O2 saturation and decrease in the partial pressure of carbon dioxide). They also downregulated the tumor necrosis factor (TNF)-α and interleukin (IL)-6 concentrations and increased the IL-10 concentrations at different times compared with time 0 and in the control group, significantly. In the CT scan, a significant decrease in the Hounsfield units and total lung volume was found by echocardiography, and in comparing the two groups, a significant difference in the parameters was noticed. The histopathology demonstrated that the BM-MSCs were able to reduce the infiltration of inflammatory cells and pulmonary hemorrhage and edema. Conclusions This study indicated that BM-MSCs play a significant role in the repair of lung injury. Electronic supplementary material The online version of this article (10.1186/s13054-018-2272-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mohammad Reza Mokhber Dezfouli
- Department of Internal Medicine, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.,Institute of Biomedical Research, University of Tehran, Tehran, Iran
| | | | - Sirous Sadeghian Chaleshtori
- Department of Internal Medicine, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran. .,Institute of Biomedical Research, University of Tehran, Tehran, Iran.
| | - Mohammad Mehdi Dehghan
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.,Institute of Biomedical Research, University of Tehran, Tehran, Iran
| | - Alireza Vajhi
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.,Institute of Biomedical Research, University of Tehran, Tehran, Iran
| | - Roshanak Mokhtari
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| |
Collapse
|
26
|
Mohammadipoor A, Antebi B, Batchinsky AI, Cancio LC. Therapeutic potential of products derived from mesenchymal stem/stromal cells in pulmonary disease. Respir Res 2018; 19:218. [PMID: 30413158 PMCID: PMC6234778 DOI: 10.1186/s12931-018-0921-x] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 10/23/2018] [Indexed: 12/15/2022] Open
Abstract
Multipotent mesenchymal stem/stromal cells (MSCs) possess robust self-renewal characteristics and the ability to differentiate into tissue-specific cells. Their therapeutic potential appears promising as evident from their efficacy in several animal models of pulmonary disorders as well as early-phase clinical trials of acute respiratory distress syndrome (ARDS) and chronic obstructive pulmonary disease (COPD). Such therapeutic efficacy might be attributed to MSC-derived products (the "secretome"), namely conditioned media (CM) and extracellular vesicles (EVs), which have been shown to play pivotal roles in the regenerative function of MSCs. Importantly, the EVs secreted by MSCs can transfer a variety of bioactive factors to modulate the function of recipient cells via various mechanisms, including ligand-receptor interactions, direct membrane fusion, endocytosis, or phagocytosis.Herein, we review the current state-of-the-science of MSC-derived CM and EVs as potential therapeutic agents in lung diseases. We suggest that the MSC-derived secretome might be an appropriate therapeutic agent for treating aggressive pulmonary disorders because of biological and logistical advantages over live cell therapy. Nonetheless, further studies are warranted to elucidate the safety and efficacy of these components in combating pulmonary diseases.
Collapse
Affiliation(s)
- Arezoo Mohammadipoor
- Multi-Organ Support Technology (MOST) Task Area, US Army Institute of Surgical Research, Fort Sam Houston, TX, USA. .,Oak Ridge Institute for Science and Education, Oak Ridge, TN, USA.
| | - Ben Antebi
- Multi-Organ Support Technology (MOST) Task Area, US Army Institute of Surgical Research, Fort Sam Houston, TX, USA
| | - Andriy I Batchinsky
- Multi-Organ Support Technology (MOST) Task Area, US Army Institute of Surgical Research, Fort Sam Houston, TX, USA.,The Geneva Foundation, Tacoma, WA, USA
| | - Leopoldo C Cancio
- Multi-Organ Support Technology (MOST) Task Area, US Army Institute of Surgical Research, Fort Sam Houston, TX, USA
| |
Collapse
|
27
|
Zhou Y, Li P, Goodwin AJ, Cook JA, Halushka PV, Chang E, Fan H. Exosomes from Endothelial Progenitor Cells Improve the Outcome of a Murine Model of Sepsis. Mol Ther 2018; 26:1375-1384. [PMID: 29599080 DOI: 10.1016/j.ymthe.2018.02.020] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/15/2018] [Accepted: 02/22/2018] [Indexed: 01/07/2023] Open
Abstract
Microvascular dysfunction leads to multi-organ failure and mortality in sepsis. Our previous studies demonstrated that administration of exogenous endothelial progenitor cells (EPCs) confers protection in sepsis as evidenced by reduced vascular leakage, improved organ function, and increased survival. We hypothesize that EPCs protect the microvasculature through the exosomes-mediated transfer of microRNAs (miRNAs). Mice were rendered septic by cecal ligation and puncture (CLP), and EPC exosomes were administered intravenously at 4 hr after CLP. EPC exosomes treatment improved survival, suppressing lung and renal vascular leakage, and reducing liver and kidney dysfunction in septic mice. EPC exosomes attenuated sepsis-induced increases in plasma levels of cytokines and chemokine. Moreover, we determined miRNA contents of EPC exosomes with next-generation sequencing and found abundant miR-126-3p and 5p. We demonstrated that exosomal miR-126-5p and 3p suppressed LPS-induced high mobility group box 1 (HMGB1) and vascular cell adhesion molecule 1 (VCAM1) levels, respectively, in human microvascular endothelial cells (HMVECs). Inhibition of microRNA-126-5p and 3p through transfection with microRNA-126-5p and 3p inhibitors abrogated the beneficial effect of EPC exosomes. The inhibition of exosomal microRNA-126 failed to block LPS-induced increase in HMGB1 and VCAM1 protein levels in HMVECs and negated the protective effect of exosomes on sepsis survival. Thus, EPC exosomes prevent microvascular dysfunction and improve sepsis outcomes potentially through the delivery of miR-126.
Collapse
Affiliation(s)
- Yue Zhou
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Biopharmaceutics, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210000, China
| | - Pengfei Li
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Andrew J Goodwin
- Department of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - James A Cook
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Perry V Halushka
- Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Pharmacology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Eugene Chang
- Department of Obstetrics-Gynecology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Hongkuan Fan
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA.
| |
Collapse
|
28
|
Sage EK, Thakrar RM, Janes SM. Genetically modified mesenchymal stromal cells in cancer therapy. Cytotherapy 2017; 18:1435-1445. [PMID: 27745603 PMCID: PMC5082580 DOI: 10.1016/j.jcyt.2016.09.003] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 08/31/2016] [Accepted: 09/02/2016] [Indexed: 12/14/2022]
Abstract
The cell therapy industry has grown rapidly over the past 3 decades, and multiple clinical trials have been performed to date covering a wide range of diseases. The most frequently used cell is mesenchymal stromal cells (MSCs), which have been used largely for their anti-inflammatory actions and in situations of tissue repair and although they have demonstrated a good safety profile, their therapeutic efficacy has been limited. In addition to these characteristics MSCs are being used for their homing and engraftment properties and have been genetically modified to enable targeted delivery of a variety of therapeutic agents in both malignant and nonmalignant conditions. This review discusses the science and technology behind genetically modified MSC therapy in malignant disease and how potential problems have been overcome to enable their use in two novel clinical trials in metastatic gastrointestinal and lung cancer.
Collapse
Affiliation(s)
- Elizabeth K Sage
- Lungs for Living Research Centre, UCL Respiratory, Rayne Institute, University College London, London, United Kingdom
| | - Ricky M Thakrar
- Lungs for Living Research Centre, UCL Respiratory, Rayne Institute, University College London, London, United Kingdom; Department of Thoracic Medicine, University College London Hospital, London, United Kingdom
| | - Sam M Janes
- Lungs for Living Research Centre, UCL Respiratory, Rayne Institute, University College London, London, United Kingdom; Department of Thoracic Medicine, University College London Hospital, London, United Kingdom.
| |
Collapse
|
29
|
Dabrowski FA, Burdzinska A, Kulesza A, Sladowska A, Zolocinska A, Gala K, Paczek L, Wielgos M. Comparison of the paracrine activity of mesenchymal stem cells derived from human umbilical cord, amniotic membrane and adipose tissue. J Obstet Gynaecol Res 2017; 43:1758-1768. [DOI: 10.1111/jog.13432] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Revised: 05/04/2017] [Accepted: 05/21/2017] [Indexed: 12/17/2022]
Affiliation(s)
- Filip A. Dabrowski
- 1st Department of Obstetrics and Gynecology; Medical University of Warsaw; Warsaw Poland
| | - Anna Burdzinska
- Department of Immunology, Transplant Medicine and Internal Diseases; Medical University of Warsaw; Warsaw Poland
| | - Agnieszka Kulesza
- Department of Immunology, Transplant Medicine and Internal Diseases; Medical University of Warsaw; Warsaw Poland
| | - Anna Sladowska
- Department of Histology and Embryology, Center for Biostructure Research; Medical University of Warsaw; Warsaw Poland
| | - Aleksandra Zolocinska
- Department of Regenerative Medicine; Maria Sklodowska-Curie Memorial Cancer Center; Warsaw Poland
| | - Kamila Gala
- Department of Immunology, Transplant Medicine and Internal Diseases; Medical University of Warsaw; Warsaw Poland
| | - Leszek Paczek
- Department of Immunology, Transplant Medicine and Internal Diseases; Medical University of Warsaw; Warsaw Poland
- Department of Bioinformatics; Institute of Biochemistry and Biophysics, Polish Academy of Sciences; Warsaw Poland
| | - Miroslaw Wielgos
- 1st Department of Obstetrics and Gynecology; Medical University of Warsaw; Warsaw Poland
| |
Collapse
|
30
|
Cruz FF, Weiss DJ, Rocco PRM. Prospects and progress in cell therapy for acute respiratory distress syndrome. Expert Opin Biol Ther 2016; 16:1353-1360. [DOI: 10.1080/14712598.2016.1218845] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
31
|
The Vascular Endothelial Growth Factors-Expressing Character of Mesenchymal Stem Cells Plays a Positive Role in Treatment of Acute Lung Injury In Vivo. Mediators Inflamm 2016; 2016:2347938. [PMID: 27313398 PMCID: PMC4895047 DOI: 10.1155/2016/2347938] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 05/03/2016] [Indexed: 12/20/2022] Open
Abstract
Recently, mesenchymal stem cells (MSC) have been proved to be beneficial in acute respiratory distress syndrome (ARDS). Vascular endothelial growth factor (VEGF) is an important angiogenesis factor that MSC release. However, the precise role of VEGF-expressing character of MSC in the MSC treatment for ARDS remains obscure. Here, we firstly knocked down the gene VEGF in MSC (MSC-ShVEGF) with lentiviral transduction. Then we injected the MSC-ShVEGF to rats with lipopolysaccharide-induced acute lung injury (ALI) via the tail vein. Data showed that MSC transplantation significantly increased VEGF levels in the lung, reduced lung permeability, protected lung endothelium from apoptosis, facilitated VE-cadherin recovery, controlled inflammation, and attenuated lung injury. However, VEGF gene knockdown in MSC led to relatively insufficient VEGF expression in the injured lung and significantly diminished the therapeutic effects of MSC on ALI, suggesting an important role of VEGF-expressing behavior of MSC in the maintenance of VEGF in the lung and the MSC treatment for ALI. Hence, we conclude that MSC restores the lung permeability and attenuates lung injury in rats with ALI in part by maintaining a “sufficient” VEGF level in the lung and the VEGF-expressing character of MSC plays a positive role in the therapeutic effects of MSC on ARDS.
Collapse
|
32
|
Hu S, Li J, Xu X, Liu A, He H, Xu J, Chen Q, Liu S, Liu L, Qiu H, Yang Y. The hepatocyte growth factor-expressing character is required for mesenchymal stem cells to protect the lung injured by lipopolysaccharide in vivo. Stem Cell Res Ther 2016; 7:66. [PMID: 27129877 PMCID: PMC4850641 DOI: 10.1186/s13287-016-0320-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 04/06/2016] [Accepted: 04/11/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) is a life-threatening condition in critically ill patients. Recently, we have found that mesenchymal stem cells (MSC) improved the permeability of human lung microvascular endothelial cells by secreting hepatocyte growth factor (HGF) in vitro. However, the properties and functions of MSC may change under complex circumstances in vivo. Here, we sought to determine the role of the HGF-expressing character of MSC in the therapeutic effects of MSC on ARDS in vivo. METHODS MSC with HGF gene knockdown (MSC-ShHGF) were constructed using lentiviral transduction. The HGF mRNA and protein levels in MSC-ShHGF were detected using quantitative real-time polymerase chain reaction and Western blotting analysis, respectively. HGF levels in the MSC culture medium were measured by enzyme-linked immunosorbent assay (ELISA). Rats with ARDS induced by lipopolysaccharide received MSC infusion via the tail vein. After 1, 6, and 24 h, rats were sacrificed. MSC retention in the lung was assessed by immunohistochemical assay. The lung wet weight to body weight ratio (LWW/BW) and Evans blue dye extravasation were obtained to reflect lung permeability. The VE-cadherin was detected with inmmunofluorescence, and the lung endothelial cell apoptosis was assessed by TUNEL assay. The severity of lung injury was evaluated using histopathology. The cytokines and HGF levels in the lung were measured by ELISA. RESULTS MSC-ShHGF with markedly lower HGF expression were successfully constructed. Treatment with MSC or MSC carrying green fluorescent protein (MSC-GFP) maintained HGF expression at relatively high levels in the lung at 24 h. MSC or MSC-GFP decreased the LWW/BW and the Evans Blue Dye extravasation, protected adherens junction VE-cadherin, and reduced the lung endothelial cell apoptosis. Furthermore, MSC or MSC-GFP reduced the inflammation and alleviated lung injury based on histopathology. However, HGF gene knockdown significantly decreased the HGF levels without any changes in the MSC retention in the lung, and diminished the protective effects of MSC on the injured lung, indicating the therapeutic effects of MSC on ARDS were partly associated with the HGF-expressing character of MSC. CONCLUSIONS MSC restores lung permeability and lung injury in part by maintaining HGF levels in the lung and the HGF-expressing character is required for MSC to protect the injured lung.
Collapse
Affiliation(s)
- Shuling Hu
- Department of Critical Care Medicine, Zhongda Hospital, Southeast University School of Medicine, No.87 Dingjiaqiao Road, Nanjing, 210009, Jiansu, P.R. China
| | - Jinze Li
- Department of Critical Care Medicine, Zhongda Hospital, Southeast University School of Medicine, No.87 Dingjiaqiao Road, Nanjing, 210009, Jiansu, P.R. China
| | - Xiuping Xu
- Department of Critical Care Medicine, Zhongda Hospital, Southeast University School of Medicine, No.87 Dingjiaqiao Road, Nanjing, 210009, Jiansu, P.R. China
| | - Airan Liu
- Department of Critical Care Medicine, Zhongda Hospital, Southeast University School of Medicine, No.87 Dingjiaqiao Road, Nanjing, 210009, Jiansu, P.R. China
| | - Hongli He
- Department of Critical Care Medicine, Zhongda Hospital, Southeast University School of Medicine, No.87 Dingjiaqiao Road, Nanjing, 210009, Jiansu, P.R. China
| | - Jingyuan Xu
- Department of Critical Care Medicine, Zhongda Hospital, Southeast University School of Medicine, No.87 Dingjiaqiao Road, Nanjing, 210009, Jiansu, P.R. China
| | - Qihong Chen
- Department of Critical Care Medicine, Zhongda Hospital, Southeast University School of Medicine, No.87 Dingjiaqiao Road, Nanjing, 210009, Jiansu, P.R. China
| | - Songqiao Liu
- Department of Critical Care Medicine, Zhongda Hospital, Southeast University School of Medicine, No.87 Dingjiaqiao Road, Nanjing, 210009, Jiansu, P.R. China
| | - Ling Liu
- Department of Critical Care Medicine, Zhongda Hospital, Southeast University School of Medicine, No.87 Dingjiaqiao Road, Nanjing, 210009, Jiansu, P.R. China
| | - Haibo Qiu
- Department of Critical Care Medicine, Zhongda Hospital, Southeast University School of Medicine, No.87 Dingjiaqiao Road, Nanjing, 210009, Jiansu, P.R. China
| | - Yi Yang
- Department of Critical Care Medicine, Zhongda Hospital, Southeast University School of Medicine, No.87 Dingjiaqiao Road, Nanjing, 210009, Jiansu, P.R. China.
| |
Collapse
|
33
|
Liu Z, Bone N, Jiang S, Park DW, Tadie JM, Deshane J, Rodriguez CA, Pittet JF, Abraham E, Zmijewski JW. AMP-Activated Protein Kinase and Glycogen Synthase Kinase 3β Modulate the Severity of Sepsis-Induced Lung Injury. Mol Med 2016; 21:937-950. [PMID: 26650187 PMCID: PMC4818252 DOI: 10.2119/molmed.2015.00198] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 11/27/2015] [Indexed: 12/29/2022] Open
Abstract
Alterations in metabolic and bioenergetic homeostasis contribute to sepsis-mediated organ injury. However, how AMP-activated protein kinase (AMPK), a major sensor and regulator of energy expenditure and production, affects development of organ injury and loss of innate capacity during polymicrobial sepsis remains unclear. In the present experiments, we found that cross-talk between the AMPK and GSK3β signaling pathways controls chemotaxis and the ability of neutrophils and macrophages to kill bacteria ex vivo. In mice with polymicrobial abdominal sepsis or more severe sepsis induced by the combination of hemorrhage and intraabdominal infection, administration of the AMPK activator metformin or the GSK3β inhibitor SB216763 reduced the severity of acute lung injury (ALI). Improved survival in metformin-treated septic mice was correlated with preservation of mitochondrial complex V (ATP synthase) function and increased amounts of ETC complex III and IV. Although immunosuppression is a consequence of sepsis, metformin effectively increased innate immune capacity to eradicate P. aeruginosa in the lungs of septic mice. We also found that AMPK activation diminished accumulation of the immunosuppressive transcriptional factor HIF-1α as well as the development of endotoxin tolerance in LPS-treated macrophages. Furthermore, AMPK-dependent preservation of mitochondrial membrane potential also prevented LPS-mediated dysfunction of neutrophil chemotaxis. These results indicate that AMPK activation reduces the severity of polymicrobial sepsis-induced lung injury and prevents the development of sepsis-associated immunosuppression.
Collapse
Affiliation(s)
- Zhongyu Liu
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Nathaniel Bone
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Shaoning Jiang
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Dae Won Park
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Jean-Marc Tadie
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Jessy Deshane
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Cilina Ann Rodriguez
- Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Jean-Francois Pittet
- Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Edward Abraham
- Office of the Dean, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Jaroslaw W Zmijewski
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| |
Collapse
|
34
|
Fujita Y, Kosaka N, Araya J, Kuwano K, Ochiya T. Extracellular vesicles in lung microenvironment and pathogenesis. Trends Mol Med 2015; 21:533-42. [PMID: 26231094 DOI: 10.1016/j.molmed.2015.07.004] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 07/01/2015] [Accepted: 07/07/2015] [Indexed: 12/18/2022]
Abstract
Increasing attention is being paid to the role of extracellular vesicles (EVs) in various lung diseases. EVs are released by a variety of cells, including respiratory cells and immune cells, and they encapsulate various molecules, such as proteins and microRNAs, as modulators of intercellular communication. Cancer cell-derived EVs play crucial roles in promoting tumor progression and modifying their microenvironment. By contrast, noncancerous cell-derived EVs demonstrate protective functions against injury, such as tissue recovery and repair, to maintain physiological homeostasis. Airway cells in contact with harmful substances may alter their EV composition and modify the balanced reciprocal interactions with surrounding mesenchymal cells. We summarize the novel findings of EV function in various lung diseases, primarily chronic obstructive pulmonary disease (COPD) and lung cancer.
Collapse
Affiliation(s)
- Yu Fujita
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1, Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 3-19-18, Nishi-shinbashi, Minato-ku, Tokyo 105-8471, Japan; Department of Pathology and Moores UCSD Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Nobuyoshi Kosaka
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1, Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; Department of Zoology, University of Oxford, The Tinbergen Building, South Parks Road, Oxford OX1 3PS, UK
| | - Jun Araya
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 3-19-18, Nishi-shinbashi, Minato-ku, Tokyo 105-8471, Japan
| | - Kazuyoshi Kuwano
- Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, 3-19-18, Nishi-shinbashi, Minato-ku, Tokyo 105-8471, Japan
| | - Takahiro Ochiya
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1, Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.
| |
Collapse
|