1
|
Li L, Song QQ, Li SR, Jia ZG, Sun XC, Zhao YT, Deng JB, Wu JJ, Ni T, Liu JS. Human umbilical cord mesenchymal stem cells-derived exosomes attenuate burn-induced acute lung injury via inhibiting ferroptosis. Acta Histochem 2024; 126:152189. [PMID: 39197328 DOI: 10.1016/j.acthis.2024.152189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024]
Abstract
Our previous study has shown that exosomes derived from human umbilical cord mesenchymal stem cells (hUCMSCs-exo) alleviated burn-induced acute lung injury (ALI). In this study, we explored a novel mechanism by which hUCMSCs-exo contributed to the inhibition of burn-induced ALI. The ALI rat model with severe burn was established for the in vivo experiments, and rats PMVECs were stimulated with the serum from burn-induced ALI rats for the in vitro experiments. The pathological changes of lung tissues were evaluated by HE staining; the cell viability was measured using CCK-8; the iron level and Fe2+ concentration were assessed using Iron Assay Kit and Fe2+ fluorescence detection probe; the mRNA expression of SLC7A11 and GPX4 were measured by qRT-PCR; the protein levels of SLC7A11, GPX4, Nrf2 and HO-1 were detected by western blot. Both the in vivo and in vitro experiments revealed that ferroptosis was significantly induced in burn-induced ALI, which as verified by increased iron level and Fe2+ concentration, and decreased SLC7A11 and GPX4 mRNA and protein levels. Furthermore, both hUCMSCs-exo and Fer-1 (the inhibitor of ferroptosis) alleviated lung inflammation and up-regulated protein levels of Nrf2 and HO-1 in the lung tissues of burn-induced ALI rats. These results suggested that hUCMSCs-exo exhibited a protective role against burn-induced ALI by inhibiting ferroptosis, partly owing to the activation of Nrf2/HO-1 pathway, thus providing a novel therapeutic strategy for burn-induced ALI.
Collapse
Affiliation(s)
- Lin Li
- Department of Burn and Plastic Surgery, The Third People's Hospital of Bengbu Affiliated to Bengbu Medical University, Bengbu 233000, China
| | - Qin-Qin Song
- Department of Burn and Plastic Surgery, The Third People's Hospital of Bengbu Affiliated to Bengbu Medical University, Bengbu 233000, China
| | - Shuang-Ru Li
- Department of Burn and Plastic Surgery, The Third People's Hospital of Bengbu Affiliated to Bengbu Medical University, Bengbu 233000, China
| | - Zhi-Gang Jia
- Department of Burn and Plastic Surgery, Affiliated Hospital of Jiangnan University, Wuxi 214028, China
| | - Xing-Chen Sun
- Department of Burn and Plastic Surgery, The Third People's Hospital of Bengbu Affiliated to Bengbu Medical University, Bengbu 233000, China
| | - Yu-Ting Zhao
- Department of Burn and Plastic Surgery, The Third People's Hospital of Bengbu Affiliated to Bengbu Medical University, Bengbu 233000, China
| | - Jia-Bin Deng
- Department of Burn and Plastic Surgery, The Third People's Hospital of Bengbu Affiliated to Bengbu Medical University, Bengbu 233000, China
| | - Jun-Jun Wu
- Department of Burn and Plastic Surgery, The Third People's Hospital of Bengbu Affiliated to Bengbu Medical University, Bengbu 233000, China
| | - Tao Ni
- Department of Burn and Plastic Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 201900, China
| | - Ji-Song Liu
- Department of Burn and Plastic Surgery, The Third People's Hospital of Bengbu Affiliated to Bengbu Medical University, Bengbu 233000, China.
| |
Collapse
|
2
|
Kumar J, Karim A, Sweety UH, Sarma H, Nurunnabi M, Narayan M. Bioinspired Approaches for Central Nervous System Targeted Gene Delivery. ACS APPLIED BIO MATERIALS 2024; 7:4975-4997. [PMID: 38100377 DOI: 10.1021/acsabm.3c00842] [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: 12/17/2023]
Abstract
Disorders of the central nervous system (CNS) which include a wide range of neurodegenerative and neurological conditions have become a serious global issue. The presence of CNS barriers poses a significant challenge to the progress of designing effective therapeutic delivery systems, limiting the effectiveness of drugs, genes, and other therapeutic agents. Natural nanocarriers present in biological systems have inspired researchers to design unique delivery systems through biomimicry. As natural resource derived delivery systems are more biocompatible, current research has been focused on the development of delivery systems inspired by bacteria, viruses, fungi, and mammalian cells. Despite their structural potential and extensive physiological function, making them an excellent choice for biomaterial engineering, the delivery of nucleic acids remains challenging due to their instability in biological systems. Similarly, the efficient delivery of genetic material within the tissues of interest remains a hurdle due to a lack of selectivity and targeting ability. Considering that gene therapies are the holy grail for intervention in diseases, including neurodegenerative disorders such as Alzheimer's disease, Parkinson's Disease, and Huntington's disease, this review centers around recent advances in bioinspired approaches to gene delivery for the prevention of CNS disorders.
Collapse
Affiliation(s)
- Jyotish Kumar
- Department of Chemistry and Biochemistry, The University of Texas at El Paso (UTEP), El Paso, Texas 79968, United States
| | - Afroz Karim
- Department of Chemistry and Biochemistry, The University of Texas at El Paso (UTEP), El Paso, Texas 79968, United States
| | - Ummy Habiba Sweety
- Environmental Science and Engineering, The University of Texas at El Paso (UTEP), El Paso, Texas 79968, United States
| | - Hemen Sarma
- Bioremediation Technology Research Group, Department of Botany, Bodoland University, Rangalikhata, Deborgaon, 783370, Kokrajhar (BTR), Assam, India
| | - Md Nurunnabi
- The Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas at El Paso, El Paso, Texas 79968, United States
| | - Mahesh Narayan
- Department of Chemistry and Biochemistry, The University of Texas at El Paso (UTEP), El Paso, Texas 79968, United States
| |
Collapse
|
3
|
Bartel S, Wolters JC, Noor H, Rafie K, Fang J, Kirchner B, Nolte-′t Hoen E, Pfaffl MW, Rutgers S, Timens W, van den Berge M, Hylkema MN. Altered Extracellular Vesicle-Derived Protein and microRNA Signatures in Bronchoalveolar Lavage Fluid from Patients with Chronic Obstructive Pulmonary Disease. Cells 2024; 13:945. [PMID: 38891077 PMCID: PMC11171984 DOI: 10.3390/cells13110945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 05/13/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive lung disease for which there is no cure. Accumulating research results suggest a role for extracellular vesicles (EVs) in the pathogenesis of COPD. This study aimed to uncover the involvement of EVs and their molecular cargo in the progression of COPD by identification of EV-associated protein and microRNA (miRNA) profiles. We isolated EVs from the bronchial alveolar lavage fluid (BALF) of 18 patients with COPD and 11 healthy controls using size-exclusion chromatography. EV isolates were characterized using nanoparticle tracking analysis and protein content. Proteomic analysis revealed a higher abundance of 284 proteins (log2FC > 1) and a lower abundance of 3 proteins (log2FC < -1) in EVs derived from patients with COPD. Ingenuity pathway analysis showed that proteins enriched in COPD-associated EVs trigger inflammatory responses, including neutrophil degranulation. Variances in surface receptors and ligands associated with COPD EVs suggest a preferential interaction with alveolar cells. Small RNAseq analysis identified a higher abundance of ten miRNAs and a lower abundance of one miRNA in EVs from COPD versus controls (Basemean > 100, FDR < 0.05). Our data indicate that the molecular composition of EVs in the BALF of patients with COPD is altered compared to healthy control EVs. Several components in COPD EVs were identified that may perpetuate inflammation and alveolar tissue destruction.
Collapse
Affiliation(s)
- Sabine Bartel
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Justina C. Wolters
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Hasnat Noor
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Karim Rafie
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9712 CP Groningen, The Netherlands
| | - Jiahua Fang
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Benedikt Kirchner
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany
- Institute of Human Genetics, LMU University Hospital, LMU Munich, 80539 Munich, Germany
| | - Esther Nolte-′t Hoen
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CS Utrecht, The Netherlands
| | - Michael W. Pfaffl
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany
| | | | - Wim Timens
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Maarten van den Berge
- Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
- Department of Pulmonary Diseases, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Machteld N. Hylkema
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
4
|
Trigo CM, Rodrigues JS, Camões SP, Solá S, Miranda JP. Mesenchymal stem cell secretome for regenerative medicine: Where do we stand? J Adv Res 2024:S2090-1232(24)00181-4. [PMID: 38729561 DOI: 10.1016/j.jare.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 02/27/2024] [Accepted: 05/03/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Mesenchymal stem cell (MSC)-based therapies have yielded beneficial effects in a broad range of preclinical models and clinical trials for human diseases. In the context of MSC transplantation, it is widely recognized that the main mechanism for the regenerative potential of MSCs is not their differentiation, with in vivo data revealing transient and low engraftment rates. Instead, MSCs therapeutic effects are mainly attributed to its secretome, i.e., paracrine factors secreted by these cells, further offering a more attractive and innovative approach due to the effectiveness and safety of a cell-free product. AIM OF REVIEW In this review, we will discuss the potential benefits of MSC-derived secretome in regenerative medicine with particular focus on respiratory, hepatic, and neurological diseases. Both free and vesicular factors of MSC secretome will be detailed. We will also address novel potential strategies capable of improving their healing potential, namely by delivering important regenerative molecules according to specific diseases and tissue needs, as well as non-clinical and clinical studies that allow us to dissect their mechanisms of action. KEY SCIENTIFIC CONCEPTS OF REVIEW MSC-derived secretome includes both soluble and non-soluble factors, organized in extracellular vesicles (EVs). Importantly, besides depending on the cell origin, the characteristics and therapeutic potential of MSC secretome is deeply influenced by external stimuli, highlighting the possibility of optimizing their characteristics through preconditioning approaches. Nevertheless, the clarity around their mechanisms of action remains ambiguous, whereas the need for standardized procedures for the successful translation of those products to the clinics urges.
Collapse
Affiliation(s)
- Catarina M Trigo
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Joana S Rodrigues
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Sérgio P Camões
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Susana Solá
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Joana P Miranda
- Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
5
|
Garcia LFC, Wowk PF, Albrecht L. Unraveling the Impact of Extracellular Vesicle-Depleted Serum on Endothelial Cell Characteristics over Time. Int J Mol Sci 2024; 25:4761. [PMID: 38731980 PMCID: PMC11084606 DOI: 10.3390/ijms25094761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/06/2024] [Accepted: 04/10/2024] [Indexed: 05/13/2024] Open
Abstract
Extracellular vesicles (EVs) are produced by all kinds of cells, including endothelial cells. It has been observed that EVs present in fetal bovine serum (FBS), broadly used in cell culture, can be a confounding factor and lead to misinterpretation of results. To investigate this phenomenon, human brain microvascular endothelial cells (HBMECs) were cultured for 2 or 24 h in the presence of EV-depleted FBS (EVdS). Cell death, gene and protein expression, and the presence of EVs isolated from these cells were evaluated. The uptake of EVs, intercellular adhesion molecule 1 (ICAM-1) expression, and monocyte adhesion to endothelial cells exposed to EVs were also evaluated. Our results revealed higher apoptosis rates in cells cultured with EVdS for 2 and 24 h. There was an increase in interleukin 8 (IL8) expression after 2 h and a decrease in interleukin 6 (IL6) and IL8 expression after 24 h of culture. Among the proteins identified in EVs isolated from cells cultured for 2 h (EV2h), several were related to ribosomes and carbon metabolism. EVs from cells cultured for 24 h (EV24h) presented a protein profile associated with cell adhesion and platelet activation. Additionally, HBMECs exhibited increased uptake of EV2h. Treatment of endothelial cells with EV2h resulted in greater ICAM-1 expression and greater adherence to monocytes than did treatment with EV24h. According to our data, HBMEC cultivated with EVdS produce EVs with different physical characteristics and protein levels that vary over time.
Collapse
Affiliation(s)
| | - Pryscilla Fanini Wowk
- Laboratório de Virologia Molecular, Instituto Carlos Chagas, Fiocruz, Curitiba 81350-010, PR, Brazil;
| | - Letusa Albrecht
- Laboratório de Pesquisa em Apicomplexa, ICC-Fiocruz-PR, Curitiba 81350-010, PR, Brazil;
| |
Collapse
|
6
|
Wei S, Lu C, Li S, Zhang Q, Cheng R, Pan S, Wu Q, Zhao X, Tian X, Zeng X, Liu Y. Efficacy and safety of mesenchymal stem cell-derived microvesicles in mouse inflammatory arthritis. Int Immunopharmacol 2024; 131:111845. [PMID: 38531171 DOI: 10.1016/j.intimp.2024.111845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 02/28/2024] [Accepted: 03/08/2024] [Indexed: 03/28/2024]
Abstract
OBJECTIVE To determine the effective and safe intravenous doses of mesenchymal stem cells (MSCs)-derived microvesicles (MVs) and to elucidate the possible causes of death in mice receiving high-dose MVs. METHODS MVs were isolated from human MSCs by gradient centrifugation. Mice with collagen-induced arthritis were treated with different doses of intravenous MVs or MSCs. Arthritis severity, white blood cell count, and serum C-reactive protein levels were measured. To assess the safety profile of MSCs and MVs, mice were treated with different doses of MSCs and MVs, and LD50 was calculated. Mouse lungs and heart were assessed by live fluorescence imaging, histopathological measurements, and immunohistochemistry to explore the possible causes of death. Serum concentrations of cTnT, cTnI, and CK-MB were determined by ELISA. With the H9C2 cardiomyocyte cell line, cellular uptake of MVs was observed using confocal microscopy and cell toxicity was assessed by CCK-8 and flow cytometry. RESULTS Intravenous treatment with MSCs and MVs alleviated inflammatory arthritis, while high doses of MSCs and MVs were lethal. Mice receiving a maximum dose of MSCs (0.1 mL of MSCs at 109/mL) died immediately, while mice receiving a maximum dose of MVs (0.1 mL of MVs at 1012/mL) exhibited tears, drooling, tachycardia, shortness of breath, unbalanced rollover, bouncing, circular crawling, mania, and death. Some mice died after exhibiting convulsions and other symptoms. All mice died shortly after injecting the maximum dose of MSCs. Histologically, mice receiving high doses of MSCs frequently developed pulmonary embolism, while those receiving high doses of MVs died of myocardial infarction. Consistently, the serum levels of cTnT, cTnI, and CK-MB were significantly increased in the MVs-treated group (P < 0.05). The LD50 of intravenous MVs was 1.60 × 1012/kg. Further, MVs could enter the cell. High doses of MVs induced cell apoptosis, though low concentrations of MVs induced cell proliferation. CONCLUSIONS Appropriate dosages of MVs and MSCs are effective treatments for inflammatory arthritis while MVs and MSCs overdose is unsafe by causing cardiopulmonary complications.
Collapse
Affiliation(s)
- Shixiong Wei
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, NO. 1 Shuai Fu Yuan, Wang Fu Jing street, Beijing 100730, China; Department of Rheumatology & Department of Rheumatology and Immunology, West China Hospital, Sichuan University, No.37 Guoxue xiang Wuhou District, Chengdu City, Sichuan Province 610041, China
| | - Chenyang Lu
- Division of Rheumatology, Department of Internal Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
| | - Sujia Li
- Department of Rheumatology & Department of Rheumatology and Immunology, West China Hospital, Sichuan University, No.37 Guoxue xiang Wuhou District, Chengdu City, Sichuan Province 610041, China
| | - Qiuping Zhang
- Department of Rheumatology & Department of Rheumatology and Immunology, West China Hospital, Sichuan University, No.37 Guoxue xiang Wuhou District, Chengdu City, Sichuan Province 610041, China
| | - Ruijuan Cheng
- Department of Rheumatology & Department of Rheumatology and Immunology, West China Hospital, Sichuan University, No.37 Guoxue xiang Wuhou District, Chengdu City, Sichuan Province 610041, China
| | - ShuYue Pan
- Department of Rheumatology & Department of Rheumatology and Immunology, West China Hospital, Sichuan University, No.37 Guoxue xiang Wuhou District, Chengdu City, Sichuan Province 610041, China
| | - QiuHong Wu
- Department of Rheumatology & Department of Rheumatology and Immunology, West China Hospital, Sichuan University, No.37 Guoxue xiang Wuhou District, Chengdu City, Sichuan Province 610041, China
| | - Xueting Zhao
- Department of Rheumatology & Department of Rheumatology and Immunology, West China Hospital, Sichuan University, No.37 Guoxue xiang Wuhou District, Chengdu City, Sichuan Province 610041, China
| | - Xinping Tian
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, NO. 1 Shuai Fu Yuan, Wang Fu Jing street, Beijing 100730, China.
| | - Xiaofeng Zeng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, NO. 1 Shuai Fu Yuan, Wang Fu Jing street, Beijing 100730, China.
| | - Yi Liu
- Department of Rheumatology & Department of Rheumatology and Immunology, West China Hospital, Sichuan University, No.37 Guoxue xiang Wuhou District, Chengdu City, Sichuan Province 610041, China.
| |
Collapse
|
7
|
Lan Q, Xiao X, Bi X, Gu Y, Ai Y. Effects of periodontal ligament stem cell-derived exosomes on osteoblastic proliferation, migration, differentiation, apoptosis, and signaling pathways. Oral Dis 2024; 30:710-718. [PMID: 36076350 DOI: 10.1111/odi.14375] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 08/24/2022] [Accepted: 09/02/2022] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Periodontitis is characterized by alveolar bone injury and absorption, with high incidence and poor treatment effect. Proliferation, migration, differentiation and apoptosis of osteoblasts are identified as key factors during the regeneration of alveolar bone tissue processes. Periodontal ligament stem cells (PDLSCs) have been proved to be a possible candidate for the treatment of periodontitis due to its multiple advantages, such as increasing the regenerative capacity of bone tissue. However, the effect of exosomes derived from PDLSCs (PDLSC-Exo) on osteoblasts remains to be further studied. METHODS AND MATERIALS In this work, cell proliferation, migration, osteogenic differentiation, and H2 O2 -induced apoptosis were detected after cells were exposed to PDLSC-Exo by CCK-8, scratch wound assay, alizarin red S and alkaline phosphatase staining, real-time PCR, flow cytometry, tunel assay, and so on. Moreover, the activation of PI3K/AKT and MEK/ERK signaling pathways was evaluated by western blotting. RESULTS We found that PDLSC-Exo are capable of promoting hFOB1.19 cell proliferation, migration and osteogenic differentiation, inhibiting H2 O2 -induced apoptosis, and activating the PI3K/AKT and MEK/ERK signaling pathways. CONCLUSION These results suggest that PDLSC-Exo may be a promising therapeutic for osteoblastic damage.
Collapse
Affiliation(s)
- Qian Lan
- Department of Periodontics, Foshan Stomatology Hospital and School of Medicine, Foshan University, Foshan, Guangdong Province, China
| | - Xin Xiao
- Department of Periodontics, Foshan Stomatology Hospital and School of Medicine, Foshan University, Foshan, Guangdong Province, China
| | - Xueting Bi
- Department of Periodontics, Foshan Stomatology Hospital and School of Medicine, Foshan University, Foshan, Guangdong Province, China
| | - Yangcong Gu
- Department of Maxillofacial Surgery, Foshan Stomatology Hospital and School of Medicine, Foshan University, Foshan, Guangdong Province, China
| | - Yilong Ai
- Department of Orthodontics, Foshan Stomatology Hospital and School of Medicine, Foshan University, Foshan, Guangdong Province, China
| |
Collapse
|
8
|
Gu Z, Sun M, Liu J, Huang Q, Wang Y, Liao J, Shu T, Tao M, Mao G, Pei Z, Meng W, Zhang X, Wei Y, Zhang S, Li S, Xiao K, Lu Y, Xu Q. Endothelium-Derived Engineered Extracellular Vesicles Protect the Pulmonary Endothelial Barrier in Acute Lung Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306156. [PMID: 38062916 PMCID: PMC10853733 DOI: 10.1002/advs.202306156] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/17/2023] [Indexed: 02/10/2024]
Abstract
Acute lung injury (ALI) is a severe respiratory disease with a high mortality rate. The integrity of the pulmonary endothelial barrier influences the development and prognosis of ALI. Therefore, it has become an important target for ALI treatment. Extracellular vesicles (EVs) are promising nanotherapeutic agents against ALI. Herein, endothelium-derived engineered extracellular vesicles (eEVs) that deliver microRNA-125b-5p (miRNA-125b) to lung tissues exerting a protective effect on endothelial barrier integrity are reported. eEVs that are modified with lung microvascular endothelial cell-targeting peptides (LET) exhibit a prolonged retention time in lung tissues and targeted lung microvascular endothelial cells in vivo and in vitro. To improve the efficacy of the EVs, miRNA-125b is loaded into EVs. Finally, LET-EVs-miRNA-125b is constructed. The results show that compared to the EVs, miRNA-125b, and EVs-miRNA-125b, LET-EVs-miRNA-125b exhibit the most significant treatment efficacy in ALI. Moreover, LET-EVs-miRNA-125b is found to have an important protective effect on endothelial barrier integrity by inhibiting cell apoptosis, promoting angiogenesis, and protecting intercellular junctions. Sequencing analysis reveals that LET-EVs-miRNA-125b downregulates early growth response-1 (EGR1) levels, which may be a potential mechanism of action. Taken together, these findings suggest that LET-EVs-miRNA-125b can treat ALI by protecting the endothelial barrier integrity.
Collapse
Affiliation(s)
- Zhengyan Gu
- Lab of Toxicology and PharmacologyFaculty of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
- Department of Pharmaceutical SciencesSchool of PharmacyNaval Medical UniversityShanghai200433P. R. China
| | - Mingxue Sun
- Lab of Toxicology and PharmacologyFaculty of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Jihao Liu
- Lab of Toxicology and PharmacologyFaculty of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Qi Huang
- School of Traditional Chinese Materia MedicaShenyang Pharmaceutical UniversityShenyang110006P. R. China
| | - Yunqin Wang
- Lab of Toxicology and PharmacologyFaculty of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Jun Liao
- Department of Pharmaceutical SciencesSchool of PharmacyNaval Medical UniversityShanghai200433P. R. China
- School of MedicineShanghai UniversityShanghai200444P. R. China
| | - Tingbin Shu
- Lab of Toxicology and PharmacologyFaculty of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Min Tao
- Lab of Toxicology and PharmacologyFaculty of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Guanchao Mao
- Lab of Toxicology and PharmacologyFaculty of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Zhipeng Pei
- Lab of Toxicology and PharmacologyFaculty of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Wenqi Meng
- Lab of Toxicology and PharmacologyFaculty of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Xinkang Zhang
- Lab of Toxicology and PharmacologyFaculty of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Youheng Wei
- State Key Laboratory of Genetic EngineeringInstitute of GeneticsFudan UniversityShanghai200433P. R. China
| | - Shanshan Zhang
- Lab of Toxicology and PharmacologyFaculty of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Songling Li
- Lab of Toxicology and PharmacologyFaculty of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| | - Kai Xiao
- Lab of Toxicology and PharmacologyFaculty of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
- Marine Biomedical Science and Technology Innovation Platform of Lingang Special AreaShanghai200433P. R. China
| | - Ying Lu
- Department of Pharmaceutical SciencesSchool of PharmacyNaval Medical UniversityShanghai200433P. R. China
| | - Qingqiang Xu
- Lab of Toxicology and PharmacologyFaculty of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
- Basic Medical Center for Pulmonary DiseaseFaculty of Naval MedicineNaval Medical UniversityShanghai200433P. R. China
| |
Collapse
|
9
|
Li L, Zhang X, Chen Y. Human Umbilical Cord Mesenchymal Stem Cell Exosome-derived miR-335-5p Alleviated Lipopolysaccharide-induced Acute Lung Injury by Regulating the m6A Level of ITGβ4 Gene. Curr Med Chem 2024; 31:5448-5467. [PMID: 38310394 DOI: 10.2174/0109298673273833231220062213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/27/2023] [Accepted: 11/16/2023] [Indexed: 02/05/2024]
Abstract
BACKGROUND Acute lung injury (ALI) is a serious complication that may accompany severe pneumonia in children. Derived from human umbilical cord mesenchymal stem cell exosome (HucMSC-Exo) can contribute to the regeneration of damaged lung tissue. This study aims to investigate the impact of HucMSC-Exo on ALI and its potential mechanisms. METHODS Firstly, RT-qPCR was performed to assess the expression of miR-335-5p. Subsequently, Pearson correlation analysis was performed to examine the correlation between METTL14 and miR-335-5p, as well as the correlation between METTL14 and ITGβ4, while RNA immunoprecipitation (RIP) was used to determine the m6A modification level of ITGβ4. Additionally, molecular biology techniques were employed to evaluate the expression of glycolysis-related factors. Definitively, an LPS-induced ALI model was established to investigate the effect of miR-335-5p on mice lung tissue. RESULTS miR-335-5p was found to be highly expressed in HucMSC-Exo. Transfection with miR-335-5p mimics resulted in increased glucose uptake. Pearson correlation analysis revealed a negative correlation between METTL14 and miR-335-5p, as well as between METTL14 and ITGβ4. The m6A level of ITGβ4 was elevated in ALI. Overexpression of METTL14 was found to reduce the expression of ITGβ4 and glucose levels, while overexpression of ITGβ4 reversed the effects of METTL14 overexpression. In vivo, results demonstrated that miR-335-5p could improve the extent of lung tissue lesions and reduce glycolytic levels. CONCLUSION This study revealed the mechanism by which miR-335-5p derived from HucMSC-Exo could alleviate LPS-induced ALI by regulating the m6A modification of ITGβ4, providing a new direction for the treatment of ALI.
Collapse
Affiliation(s)
- Linrui Li
- Department of Respiratory Medicine, Hunan Children's Hospital, Changsha, 410006, China
| | - Xi Zhang
- Department of Respiratory Medicine, Hunan Children's Hospital, Changsha, 410006, China
| | - Yanping Chen
- Department of Respiratory Medicine, Hunan Children's Hospital, Changsha, 410006, China
| |
Collapse
|
10
|
Zhang C, Li X, Gao D, Zhu H, Wang S, Tan B, Yang A. Network Pharmacology and Experimental Validation of the Anti-Inflammatory Effect of Tingli Dazao Xiefei Decoction in Acute Lung Injury Treatment. J Inflamm Res 2023; 16:6195-6209. [PMID: 38145012 PMCID: PMC10748588 DOI: 10.2147/jir.s433840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 12/12/2023] [Indexed: 12/26/2023] Open
Abstract
Purpose Tingli Dazao Xiefei Decoction (TDXD) is a Traditional Chinese Medicine (TCM) formula used to treat acute lung injury (ALI). However, the precise mechanism of TDXD in treating ALI remains unclear. We investigated the therapeutic mechanism of TDXD against ALI using a complementary approach combining network pharmacology, molecular docking, and in vitro and in vivo experiments. Material and Methods Potential drug targets of TDXD and relevant target genes associated with ALI were retrieved from Chinese medicines and disease genes databases. Bioinformatics technology was employed to screen potential active ingredients and core targets. Validation experiments were conducted using a lipopolysaccharide (LPS)-induced ALI mouse (C57BL/6J) model, LPS-induced inflammatory RAW264.7 cells, and molecular docking between active compounds of TDXD and potential targets. Results Network pharmacology suggested that the mechanism of TDXD against ALI involved phosphoinositide 3-kinase (PI3K) / protein kinase B (AKT) / phosphatase and tensin homolog (PTEN) and Janus kinase 2 (JAK2) / signal transducer and activator of transcription 3 (STAT3) pathways. Quercetin, β-sitosterol, kaempferol, isorhamnetin, and L-stepholidine were identified as the main active compounds of TDXD that exerted anti-ALI effects. Molecular docking indicated that these compounds exhibited good binding capabilities (≤ -5kcal/mol) to key targets in PI3K/AKT/PTEN and JAK2/STAT3 signaling pathways. In the animal model, TDXD alleviated injuries and inflammatory responses in lung tissues, accompanied by inhibition of expression of tumor necrosis factor-α (TNF-α), Interleukin-6 (IL-6), STAT3, and Suppressor of Cytokine Signaling 3 (SOCS3) mRNA, and key proteins in PI3K/AKT/PTEN and JAK2/STAT3 pathways (all P values < 0.05). Cell based experiments showed that TDXD dose-dependently inhibited the expression of essential proteins in PI3K/AKT/PTEN and JAK2/STAT3 pathways (P < 0.05). Conclusion This study revealed that the mechanism of TDXD in ALI treatment might involve simultaneous regulation of PI3K/AKT/PTEN and JAK2/STAT3 pathways.
Collapse
Affiliation(s)
- Chengxi Zhang
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Center for Traditional Chinese Medicine and Epidemic Disease, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, People’s Republic of China
| | - Xiaoqian Li
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Center for Traditional Chinese Medicine and Epidemic Disease, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, People’s Republic of China
| | - Dan Gao
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Center for Traditional Chinese Medicine and Epidemic Disease, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, People’s Republic of China
| | - Huahe Zhu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Shun Wang
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Center for Traditional Chinese Medicine and Epidemic Disease, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, People’s Republic of China
| | - Bo Tan
- Laboratory of Clinical Pharmacokinetics, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Aidong Yang
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Center for Traditional Chinese Medicine and Epidemic Disease, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, People’s Republic of China
| |
Collapse
|
11
|
Didamoony MA, Soubh AA, Atwa AM, Ahmed LA. Innovative preconditioning strategies for improving the therapeutic efficacy of extracellular vesicles derived from mesenchymal stem cells in gastrointestinal diseases. Inflammopharmacology 2023; 31:2973-2993. [PMID: 37874430 PMCID: PMC10692273 DOI: 10.1007/s10787-023-01350-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 09/20/2023] [Indexed: 10/25/2023]
Abstract
Gastrointestinal (GI) diseases have become a global health issue and an economic burden due to their wide distribution, late prognosis, and the inefficacy of recent available medications. Therefore, it is crucial to search for new strategies for their management. In the recent decades, mesenchymal stem cells (MSCs) therapy has attracted attention as a viable option for treating a myriad of GI disorders such as hepatic fibrosis (HF), ulcerative colitis (UC), acute liver injury (ALI), and non-alcoholic fatty liver disease (NAFLD) due to their regenerative and paracrine properties. Importantly, recent studies have shown that MSC-derived extracellular vesicles (MSC-EVs) are responsible for most of the therapeutic effects of MSCs. In addition, EVs have revealed several benefits over their parent MSCs, such as being less immunogenic, having a lower risk of tumour formation, being able to cross biological barriers, and being easier to store. MSC-EVs exhibited regenerative, anti-oxidant, anti-inflammatory, anti-apoptotic, and anti-fibrotic effects in different experimental models of GI diseases. However, a key issue with their clinical application is the maintenance of their stability and efficacy following in vivo transplantation. Preconditioning of MSC-EVs or their parent cells is one of the novel methods used to improve their effectiveness and stability. Herein, we discuss the application of MSC-EVs in several GI disorders taking into account their mechanism of action. We also summarise the challenges and restrictions that need to be overcome to promote their clinical application in the treatment of various GI diseases as well as the recent developments to improve their effectiveness. A representation of the innovative preconditioning techniques that have been suggested for improving the therapeutic efficacy of MSC-EVs in GI diseases. The pathological conditions in various GI disorders (ALI, UC, HF and NAFLD) create a harsh environment for EVs and their parents, increasing the risk of apoptosis and senescence of MSCs and thereby diminishing MSC-EVs yield and restricting their large-scale applications. Preconditioning with pharmacological agents or biological mediators can improve the therapeutic efficacy of MSC-EVs through their adaption to the lethal environment to which they are subjected. This can result in establishment of a more conducive environment and activation of numerous vital trajectories that act to improve the immunomodulatory, reparative and regenerative activities of the derived EVs, as a part of MSCs paracrine system. ALI, acute liver injury; GI diseases, gastrointestinal diseases; HF, hepatic fibrosis; HSP, heat shock protein; miRNA, microRNA; mRNA, messenger RNA; MSC-EVs, mesenchymal stem cell-derived extracellular vesicles; NAFLD, non-alcoholic fatty liver disease; UC, ulcerative colitis.
Collapse
Affiliation(s)
- Manar A Didamoony
- Faculty of Pharmacy, Pharmacology and Toxicology Department, Egyptian Russian University, Cairo, 11829, Egypt.
| | - Ayman A Soubh
- Faculty of Pharmacy, Pharmacology and Toxicology Department, Ahram Canadian University, 4th Industrial Zone, Banks Complex, 6th of October City, Giza, 12451, Egypt
| | - Ahmed M Atwa
- Faculty of Pharmacy, Pharmacology and Toxicology Department, Egyptian Russian University, Cairo, 11829, Egypt
| | - Lamiaa A Ahmed
- Faculty of Pharmacy, Pharmacology and Toxicology Department, Cairo University, Cairo, 11562, Egypt.
| |
Collapse
|
12
|
Fu H, Liang X, Tan W, Hu X. Unraveling the protective mechanisms of Chuanfangyihao against acute lung injury: Insights from experimental validation. Exp Ther Med 2023; 26:535. [PMID: 37869635 PMCID: PMC10587870 DOI: 10.3892/etm.2023.12234] [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: 06/25/2023] [Accepted: 08/21/2023] [Indexed: 10/24/2023] Open
Abstract
Chuanfangyihao (CFYH) is an effective treatment for acute lung injury (ALI) in clinical practice; however, its underlying mechanism of action remains unclear. Therefore, the aim of the present study was to elucidate the pharmacological mechanism of action of CFYH in ALI through experimental validation. First, a rat model of ALI was established using lipopolysaccharide (LPS). Next, the pathological changes in the lungs of the rats and the pathological damage were scored. The wet/dry weight ratios were measured, and ROS content was detected using flow cytometry. ELISA was used to examine IL-6, TNF-α, IL-1β, IL-18, and LDH levels. Immunohistochemistry was used to detect Beclin-1 and NLRP3 expression. Western blotting was performed to analyze the expression of HMGB1, RAGE, TLR4, NF-κB p65, AMPK, p-AMPK, mTOR, p-mTOR, Beclin-1, LC3-II/I, p62, Bcl-2, Bax, Caspase-3, Caspase-1, and GSDMD-NT. The mRNA levels of HMGB1, RAGE, AMPK, mTOR, and HIF-1α were determined using reverse transcription quantitative PCR. CFYH alleviated pulmonary edema and decreased the expression of IL-6, TNF-α, TLR4, NF-κB p65, HMGB1/RAGE, ROS, and HIF-1α. In addition, pretreatment with CFYH reversed ALI-induced programmed cell death. In conclusion, CFYH alleviates LPS-induced ALI, and these findings provide a preliminary clarification of the predominant mechanism of action of CFYH in ALI.
Collapse
Affiliation(s)
- Hongfang Fu
- Infectious Disease Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| | - Xiao Liang
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| | - Wanying Tan
- Infectious Disease Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| | - Xiaoyu Hu
- Infectious Disease Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| |
Collapse
|
13
|
Wang L, Li Z, Lu T, Su L, Mao C, Zhang Y, Zhang X, Jiang X, Xie H, Yu X. The potential mechanism of Choulingdan mixture in improving acute lung injury based on HPLC-Q-TOF-MS, network pharmacology and in vivo experiments. Biomed Chromatogr 2023; 37:e5709. [PMID: 37533317 DOI: 10.1002/bmc.5709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 06/29/2023] [Accepted: 07/07/2023] [Indexed: 08/04/2023]
Abstract
Choulingdan mixture (CLDM) is an empirical clinical prescription for the adjuvant treatment of acute lung injury (ALI). CLDM has been used for almost 30 years in the clinic. However, its mechanism for improving ALI still needs to be investigated. In this study, high-performance liquid chromatography-quadrupole/time-of-flight mass spectrometry (HPLC-Q-TOF-MS/MS) was applied to characterize the overall chemical composition of CLDM. A total of 93 ingredients were characterized, including 25 flavonoids, 20 organic acids, 11 saponins, nine terpenoids, seven tannins and 21 other compounds. Then network pharmacology was applied to predict the potential bioactive components, target genes and signaling pathways of CLDM in improving ALI. Additionally, molecular docking was performed to demonstrate the interaction between the active ingredients and the disease targets. Finally, animal experiments further confirmed that CLDM significantly inhibits pulmonary inflammation, pulmonary edema and oxidative stress in lipopolysaccharide-induced ALI mice by inhibiting the PI3K-AKT signaling pathway. This study enhanced the amount and accuracy of compounds of CLDM and provided new insights into CLDM preventing and treating ALI.
Collapse
Affiliation(s)
- Lili Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhengyan Li
- Department of Pharmacy, Kunming Municipal Hospital of Traditional Chinese Medicine, Kunming, China
| | - Tulin Lu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lianlin Su
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chunqin Mao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yiting Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xinrui Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaofeng Jiang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hui Xie
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaoling Yu
- Department of Pharmacy, Kunming Municipal Hospital of Traditional Chinese Medicine, Kunming, China
| |
Collapse
|
14
|
Zhuang X, Jiang Y, Yang X, Fu L, Luo L, Dong Z, Zhao J, Hei F. Advances of mesenchymal stem cells and their derived extracellular vesicles as a promising therapy for acute respiratory distress syndrome: from bench to clinic. Front Immunol 2023; 14:1244930. [PMID: 37711624 PMCID: PMC10497773 DOI: 10.3389/fimmu.2023.1244930] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is an acute inflammatory lung injury characterized by diffuse alveolar damage. The period prevalence of ARDS was 10.4% of ICU admissions in 50 countries. Although great progress has been made in supportive care, the hospital mortality rate of severe ARDS is still up to 46.1%. Moreover, up to now, there is no effective pharmacotherapy for ARDS and most clinical trials focusing on consistently effective drugs have met disappointing results. Mesenchymal stem cells (MSCs) and their derived extracellular vesicles (EVs) have spawned intense interest of a wide range of researchers and clinicians due to their robust anti-inflammatory, anti-apoptotic and tissue regeneration properties. A growing body of evidence from preclinical studies confirmed the promising therapeutic potential of MSCs and their EVs in the treatment of ARDS. Based on the inspiring experimental results, clinical trials have been designed to evaluate safety and efficacy of MSCs and their EVs in ARDS patients. Moreover, trials exploring their optimal time window and regimen of drug administration are ongoing. Therefore, this review aims to present an overview of the characteristics of mesenchymal stem cells and their derived EVs, therapeutic mechanisms for ARDS and research progress that has been made over the past 5 years.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Feilong Hei
- Department of Cardiopulmonary Bypass, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
15
|
Zhuang C, Kang M, Lee M. Delivery systems of therapeutic nucleic acids for the treatment of acute lung injury/acute respiratory distress syndrome. J Control Release 2023; 360:1-14. [PMID: 37330013 DOI: 10.1016/j.jconrel.2023.06.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 05/10/2023] [Accepted: 06/12/2023] [Indexed: 06/19/2023]
Abstract
Acute lung injury (ALI)/ acute respiratory distress syndrome (ARDS) is a devastating inflammatory lung disease with a high mortality rate. ALI/ARDS is induced by various causes, including sepsis, infections, thoracic trauma, and inhalation of toxic reagents. Corona virus infection disease-19 (COVID-19) is also a major cause of ALI/ARDS. ALI/ARDS is characterized by inflammatory injury and increased vascular permeability, resulting in lung edema and hypoxemia. Currently available treatments for ALI/ARDS are limited, but do include mechanical ventilation for gas exchange and treatments supportive of reduction of severe symptoms. Anti-inflammatory drugs such as corticosteroids have been suggested, but their clinical effects are controversial with possible side-effects. Therefore, novel treatment modalities have been developed for ALI/ARDS, including therapeutic nucleic acids. Two classes of therapeutic nucleic acids are in use. The first constitutes knock-in genes for encoding therapeutic proteins such as heme oxygenase-1 (HO-1) and adiponectin (APN) at the site of disease. The other is oligonucleotides such as small interfering RNAs and antisense oligonucleotides for knock-down expression of target genes. Carriers have been developed for efficient delivery for therapeutic nucleic acids into the lungs based on the characteristics of the nucleic acids, administration routes, and targeting cells. In this review, ALI/ARDS gene therapy is discussed mainly in terms of delivery systems. The pathophysiology of ALI/ARDS, therapeutic genes, and their delivery strategies are presented for development of ALI/ARDS gene therapy. The current progress suggests that selected and appropriate delivery systems of therapeutic nucleic acids into the lungs may be useful for the treatment of ALI/ARDS.
Collapse
Affiliation(s)
- Chuanyu Zhuang
- Department of Bioengineering, College of Engineering, Hanyang University, Wangsimni-ro 222, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Minji Kang
- Department of Bioengineering, College of Engineering, Hanyang University, Wangsimni-ro 222, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Minhyung Lee
- Department of Bioengineering, College of Engineering, Hanyang University, Wangsimni-ro 222, Seongdong-gu, Seoul 04763, Republic of Korea.
| |
Collapse
|
16
|
Li Y, Zhang C, Zhao Z. CircSLCO3A1 depletion ameliorates lipopolysaccharide-induced inflammation and apoptosis of human pulmonary alveolar epithelial cells through the miR-424-5p/HMGB3 pathway. Mol Cell Toxicol 2023:1-12. [PMID: 37359246 PMCID: PMC10211294 DOI: 10.1007/s13273-023-00341-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2023] [Indexed: 06/28/2023]
Abstract
Background Recent studies have shown the pathogenesis of acute lung injury (ALI) involves circular RNA (circRNA). However, there are no data on the role of circSLCO3A1 in ALI and the underlying mechanism. Objective ALI-like cell injury was induced by stimulating human pulmonary alveolar epithelial cells (HPAEpiCs) using lipopolysaccharide (LPS). The expression of circSLCO3A1, miR-424-5p and high mobility group box 3 (HMGB3) was detected by quantitative real-time polymerase chain reaction. Cell viability and cell apoptosis were assessed by cell counting kit-8 (CCK-8) assay and flow cytometry analysis, respectively. Enzyme-linked immunosorbent assay was performed to determine the production of interleukin-1β (IL-1β), IL-6, tumor necrosis factor-α (TNF-α) and monocyte chemotactic protein 1 (MCP-1). Caspase-3 activity was detected by caspase-3 activity assay. Protein expression of inducible NOS (iNOS), cyclooxygenase-2 (COX2), p-p65 and p65 was analyzed by Western blot. The interactions among circSLCO3A1, miR-424-5p and HMGB3 were identified by dual-luciferase reporter assay, RNA immunoprecipitation assay and RNA pull-down assay. Results CircSLCO3A1 and HMGB3 expression were significantly increased, while miR-424-5p was decreased in LPS-treated HPAEpiCs and the serum of septic ALI patients in comparison with controls. CircSLCO3A1 knockdown assuaged LPS-induced HPAEpiC inflammation and apoptosis. Besides, circSLCO3A1 targeted miR-424-5p and regulated LPS-triggered HPAEpiC inflammation and apoptosis by binding to miR-424-5p. Under the treatment of LPS, miR-424-5p mediated HPAEpiC disorders by targeting HMGB3. Importantly, circSLCO3A1 modulated HMGB3 production by interacting with miR-424-5p. Conclusion CircSLCO3A1 absence assuaged LPS-induced HPAEpiC inflammation and apoptosis through the miR-424-5p/HMGB3 axis. Highlights CircSLCO3A1 expression was upregulated in LPS-induced HPAEpiCs and sepsis-induced ALI patients.CircSLCO3A1 depletion protected against LPS-induced HPAEpiC disorders.CircSLCO3A1 bound to miR-424-5p in HPAEpiCs.MiR-424-5p targeted HMGB3 in HPAEpiCs.CircSLCO3A1 regulated HMGB3 expression through miR-424-5p. Supplementary Information The online version contains supplementary material available at 10.1007/s13273-023-00341-6.
Collapse
Affiliation(s)
- Yan Li
- Department of Emergency Medicine, China-Japan Union Hospital of Jilin University, Changchun, Jilin China
| | - Chunmei Zhang
- Department of Critical Medicine, China-Japan Union Hospital of Jilin University, No. 126, Xiantai Street, Changchun, 130033 Jilin China
| | - Zhongyan Zhao
- Department of Critical Medicine, China-Japan Union Hospital of Jilin University, No. 126, Xiantai Street, Changchun, 130033 Jilin China
| |
Collapse
|
17
|
Homma K, Bazhanov N, Hashimoto K, Shimizu M, Heathman T, Hao Q, Nawgiri R, Muthukumarana V, Lee JW, Prough DS, Enkhbaatar P. Mesenchymal stem cell-derived exosomes for treatment of sepsis. Front Immunol 2023; 14:1136964. [PMID: 37180159 PMCID: PMC10169690 DOI: 10.3389/fimmu.2023.1136964] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 03/31/2023] [Indexed: 05/15/2023] Open
Abstract
Introduction The pathogenesis of sepsis is an imbalance between pro-inflammatory and anti-inflammatory responses. At the onset of sepsis, the lungs are severely affected, and the injury progresses to acute respiratory distress syndrome (ARDS), with a mortality rate of up to 40%. Currently, there is no effective treatment for sepsis. Cellular therapies using mesenchymal stem cells (MSCs) have been initiated in clinical trials for both ARDS and sepsis based on a wealth of pre-clinical data. However, there remains concern that MSCs may pose a tumor risk when administered to patients. Recent pre-clinical studies have demonstrated the beneficial effects of MSC-derived extracellular vesicles (EVs) for the treatment of acute lung injury (ALI) and sepsis. Methods After recovery of initial surgical preparation, pneumonia/sepsis was induced in 14 adult female sheep by the instillation of Pseudomonas aeruginosa (~1.0×1011 CFU) into the lungs by bronchoscope under anesthesia and analgesia. After the injury, sheep were mechanically ventilated and continuously monitored for 24 h in a conscious state in an ICU setting. After the injury, sheep were randomly allocated into two groups: Control, septic sheep treated with vehicle, n=7; and Treatment, septic sheep treated with MSC-EVs, n=7. MSC-EVs infusions (4ml) were given intravenously one hour after the injury. Results The infusion of MSCs-EVs was well tolerated without adverse events. PaO2/FiO2 ratio in the treatment group tended to be higher than the control from 6 to 21 h after the lung injury, with no significant differences between the groups. No significant differences were found between the two groups in other pulmonary functions. Although vasopressor requirement in the treatment group tended to be lower than in the control, the net fluid balance was similarly increased in both groups as the severity of sepsis progressed. The variables reflecting microvascular hyperpermeability were comparable in both groups. Conclusion We have previously demonstrated the beneficial effects of bone marrow-derived MSCs (10×106 cells/kg) in the same model of sepsis. However, despite some improvement in pulmonary gas exchange, the present study demonstrated that EVs isolated from the same amount of bone marrow-derived MSCs failed to attenuate the severity of multiorgan dysfunctions.
Collapse
Affiliation(s)
- Kento Homma
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, United States
| | - Nikolay Bazhanov
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, United States
| | - Kazuki Hashimoto
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, United States
| | - Masaru Shimizu
- Department of Anesthesiology, University of California, San Francisco, CA, United States
| | - Thomas Heathman
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, United States
| | - Qi Hao
- Department of Anesthesiology, University of California, San Francisco, CA, United States
| | - Ranjana Nawgiri
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Vidarshi Muthukumarana
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Jae Woo Lee
- Department of Anesthesiology, University of California, San Francisco, CA, United States
| | - Donald S. Prough
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, United States
| | - Perenlei Enkhbaatar
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
18
|
Effect of Hyaluronic Acid and Mesenchymal Stem Cells Secretome Combination in Promoting Alveolar Regeneration. Int J Mol Sci 2023; 24:ijms24043642. [PMID: 36835068 PMCID: PMC9966269 DOI: 10.3390/ijms24043642] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/07/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
Pharmacological therapies in lung diseases are nowadays useful in reducing the symptomatology of lung injury. However, they have not yet been translated to effective treatment options able to restore the lung tissue damage. Cell-therapy based on Mesenchymal Stem Cells (MSCs) is an attractive, as well as new therapeutic approach, although some limitations can be ascribed for therapeutic use, such as tumorigenicity and immune rejection. However, MSCs have the capacity to secrete multiple paracrine factors, namely secretome, capable of regulating endothelial and epithelial permeability, decrease inflammation, enhancing tissue repair, and inhibiting bacterial growth. Furthermore, Hyaluronic acid (HA) has been demonstrated to have particularly efficacy in promoting the differentiation of MSCs in Alveolar type II (ATII) cells. In this frame, the combination of HA and secretome to achieve the lung tissue regeneration has been investigated for the first time in this work. Overall results showed how the combination of HA (low and medium molecular weight HA) plus secretome could enhance MSCs differentiation in ATII cells (SPC marker expression of about 5 ng/mL) compared to the only HA or secretome solutions alone (SPC about 3 ng/mL, respectively). Likewise, cell viability and cell rate of migration were reported to be improved for HA and secretome blends, indicating an interesting potentiality of such systems for lung tissue repair. Moreover, an anti-inflammatory profile has been revealed when dealing with HA and secretome mixtures. Therefore, these promising results can allow important advance in the accomplishment of the future therapeutic approach in respiratory diseases, up to date still missing.
Collapse
|
19
|
Liu J, Xing F, Fu Q, He B, Jia Z, Du J, Li Y, Zhang X, Chen X. hUC-MSCs exosomal miR-451 alleviated acute lung injury by modulating macrophage M2 polarization via regulating MIF-PI3K-AKT signaling pathway. ENVIRONMENTAL TOXICOLOGY 2022; 37:2819-2831. [PMID: 35997581 DOI: 10.1002/tox.23639] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 08/01/2022] [Accepted: 08/04/2022] [Indexed: 06/15/2023]
Abstract
In the previous study, we have proved that exosomal miR-451 from human umbilical cord mesenchymal stem cells (hUC-MSCs) attenuated burn-induced acute lung injury (ALI). However, the mechanism of exosomal miR-451 in ALI remains unclear. Therefore, this study aimed to study the molecular mechanism of hUC-MSCs-derived exosomal miR-451 on ALI by regulating macrophage polarization. Exosomes were isolated and identified by transmission electron microscope (TEM) and nanoparticle tracking analysis (NTA). The expression of miR-451, macrophage migration inhibitory factor (MIF) and PI3K/AKT signaling pathway proteins were detected by qRT-PCR and western blot. Flow cytometry was used to detect the CD80 and CD206 positive cells. Severe burn rat model was established and HE was used to detect the inflammatory cell infiltration and inflammatory injury. Dual luciferase reporter system was used to detect the regulation of miR-451 to MIF. The contents of cytokines were detected by ELISA. The results showed that hUC-MSCs exosomes promoted macrophage M1 to M2 polarization. Furthermore, hUC-MSCs-derived exosomal miR-451 alleviated ALI development and promoted macrophage M1 to M2 polarization. Moreover, MIF was a direct target of miR-451. Downregulation of MIF regulated by miR-451 alleviated ALI development promoted macrophage M1 to M2 polarization. In addition, we found that MIF and hUC-MSCs-derived exosomal miR-451 participated in ALI by regulating PI3K/AKT signaling pathway. In conclusion, we indicated that hUC-MSCs-derived exosomal miR-451 alleviated ALI by modulating macrophage M2 polarization via regulating MIF-PI3K-AKT signaling pathway, which provided great scientific significance and clinical application value for the treatment of burn-induced ALI.
Collapse
Affiliation(s)
- Jisong Liu
- Department of Burn and Plastic Surgery, Third Hospital of Bengbu, Bengbu, Anhui, China
| | - Fuxi Xing
- Department of Burn and Plastic Surgery, Third Hospital of Bengbu, Bengbu, Anhui, China
| | - Quanyou Fu
- Department of Burn and Plastic Surgery, Third Hospital of Bengbu, Bengbu, Anhui, China
| | - Bo He
- Department of Burn and Plastic Surgery, Third Hospital of Bengbu, Bengbu, Anhui, China
| | - Zhigang Jia
- Department of Burn and Plastic Surgery, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Juan Du
- Department of Minimally Invasive Surgery, Third Hospital of Bengbu, Bengbu, Anhui, China
| | - Yong Li
- Department of Burn and Plastic Surgery, Third Hospital of Bengbu, Bengbu, Anhui, China
| | - Xiangzhou Zhang
- Department of Burn and Plastic Surgery, Third Hospital of Bengbu, Bengbu, Anhui, China
| | - Xulin Chen
- Department of Burns, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
20
|
Hu Q, Zhang S, Yang Y, Yao JQ, Tang WF, Lyon CJ, Hu TY, Wan MH. Extracellular vesicles in the pathogenesis and treatment of acute lung injury. Mil Med Res 2022; 9:61. [PMID: 36316787 PMCID: PMC9623953 DOI: 10.1186/s40779-022-00417-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 09/19/2022] [Indexed: 11/05/2022] Open
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are common life-threatening lung diseases associated with acute and severe inflammation. Both have high mortality rates, and despite decades of research on clinical ALI/ARDS, there are no effective therapeutic strategies. Disruption of alveolar-capillary barrier integrity or activation of inflammatory responses leads to lung inflammation and injury. Recently, studies on the role of extracellular vesicles (EVs) in regulating normal and pathophysiologic cell activities, including inflammation and injury responses, have attracted attention. Injured and dysfunctional cells often secrete EVs into serum or bronchoalveolar lavage fluid with altered cargoes, which can be used to diagnose and predict the development of ALI/ARDS. EVs secreted by mesenchymal stem cells can also attenuate inflammatory reactions associated with cell dysfunction and injury to preserve or restore cell function, and thereby promote cell proliferation and tissue regeneration. This review focuses on the roles of EVs in the pathogenesis of pulmonary inflammation, particularly ALI/ARDS.
Collapse
Affiliation(s)
- Qian Hu
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Shu Zhang
- Department of Emergency Medicine, Emergency Medical Laboratory, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Yue Yang
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Jia-Qi Yao
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Wen-Fu Tang
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Christopher J Lyon
- Center of Cellular and Molecular Diagnosis, Tulane University School of Medicine, 1430 Tulane Ave., New Orleans, LA, 70112, USA.,Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, 1430 Tulane Ave., New Orleans, LA, 70112, USA
| | - Tony Ye Hu
- Center of Cellular and Molecular Diagnosis, Tulane University School of Medicine, 1430 Tulane Ave., New Orleans, LA, 70112, USA. .,Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, 1430 Tulane Ave., New Orleans, LA, 70112, USA.
| | - Mei-Hua Wan
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China. .,West China Hospital (Airport) of Sichuan University, Chengdu, 610299, China.
| |
Collapse
|
21
|
McLaughlin C, Datta P, Singh YP, Lo A, Horchler S, Elcheva IA, Ozbolat IT, Ravnic DJ, Koduru SV. Mesenchymal Stem Cell-Derived Extracellular Vesicles for Therapeutic Use and in Bioengineering Applications. Cells 2022; 11:3366. [PMID: 36359762 PMCID: PMC9657427 DOI: 10.3390/cells11213366] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/10/2022] [Accepted: 10/19/2022] [Indexed: 07/25/2023] Open
Abstract
Extracellular vesicles (EVs) are small lipid bilayer-delimited particles that are naturally released from cells into body fluids, and therefore can travel and convey regulatory functions in the distal parts of the body. EVs can transmit paracrine signaling by carrying over cytokines, chemokines, growth factors, interleukins (ILs), transcription factors, and nucleic acids such as DNA, mRNAs, microRNAs, piRNAs, lncRNAs, sn/snoRNAs, mtRNAs and circRNAs; these EVs travel to predecided destinations to perform their functions. While mesenchymal stem cells (MSCs) have been shown to improve healing and facilitate treatments of various diseases, the allogenic use of these cells is often accompanied by serious adverse effects after transplantation. MSC-produced EVs are less immunogenic and can serve as an alternative to cellular therapies by transmitting signaling or delivering biomaterials to diseased areas of the body. This review article is focused on understanding the properties of EVs derived from different types of MSCs and MSC-EV-based therapeutic options. The potential of modern technologies such as 3D bioprinting to advance EV-based therapies is also discussed.
Collapse
Affiliation(s)
- Caroline McLaughlin
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Pallab Datta
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER) Kolkata, West Bengal 700054, India
| | - Yogendra P. Singh
- Department of Biomedical Engineering, Materials Research Institute, The Huck Institutes of Life Sciences, Penn State University, University Park, PA 16802, USA
| | - Alexis Lo
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Summer Horchler
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Irina A. Elcheva
- Department of Pediatrics, Hematology/Oncology, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Ibrahim T. Ozbolat
- Department of Biomedical Engineering, Materials Research Institute, The Huck Institutes of Life Sciences, Penn State University, University Park, PA 16802, USA
| | - Dino J. Ravnic
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Srinivas V. Koduru
- Department of Surgery, Penn State Health Milton S. Hershey Medical Center, Hershey, PA 17033, USA
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
22
|
Extracellular Vesicles Derived from Mesenchymal Stem Cells: A Potential Biodrug for Acute Respiratory Distress Syndrome Treatment. BioDrugs 2022; 36:701-715. [PMID: 36087245 PMCID: PMC9463673 DOI: 10.1007/s40259-022-00555-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2022] [Indexed: 12/15/2022]
Abstract
Acute respiratory distress syndrome (ARDS) is a severe respiratory disease associated with high morbidity and mortality in the clinic. In the face of limited treatment options for ARDS, extracellular vesicles derived from mesenchymal stem cells (MSC-EVs) have recently shown promise. They regulate levels of growth factors, cytokines, and other internal therapeutic molecules. The possible therapeutic mechanisms of MSC-EVs include anti-inflammatory, cell injury repair, alveolar fluid clearance, and microbe clearance. The potent therapeutic ability and biocompatibility of MSC-EVs have enabled them as an alternative option to ameliorate ARDS. In this review, recent advances, therapeutic mechanisms, advantages and limitations, as well as improvements of using MSC-EVs to treat ARDS are summarized. This review is expected to provide a brief view of the potential applications of MSC-EVs as novel biodrugs to treat ARDS.
Collapse
|
23
|
Wang X, Feng J, Dai H, Mo J, Luo B, Luo C, Zhang W, Pan L. microRNA-130b-3p delivery by mesenchymal stem cells-derived exosomes confers protection on acute lung injury. Autoimmunity 2022; 55:597-607. [PMID: 36018063 DOI: 10.1080/08916934.2022.2094370] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
OBJECTIVE Researchers have investigated miR-130b-3p in lung disease pathology, such as lung fibrosis. The present study was performed to elucidate the miR-130b-3p-involved mechanism in acute lung injury (ALI) through delivery by mesenchymal stem cells-derived exosomes (MSCs-Exo). METHODS ALI mouse models were induced via intratracheal administration of lipopolysaccharide (LPS) and treated with MSCs-Exo. Lung dry-wet (W/D) ratio, inflammatory factors in the bronchoalveolar lavage fluid, pathological damage and apoptosis in the lung tissues were analyzed. Expression levels of miR-130b-3p and TGFBR1 were measured in the mouse lung tissues, and the interaction between miR-130b-3p and TGFBR1 was studied. RESULTS MSCs-Exo relieved LPS-induced ALI in mice by reducing lung W/D ratio and inflammatory response, and attenuating lung tissue pathological damage and reducing the alveolar cell apoptosis. miR-130b-3p delivery by MSCs-Exo reduced LPS-induced ALI in mice. TGFBR1 was determined to be a downstream target gene of miR-130b-3p. Inhibition of TGFBR1 could remit LPS-induced ALI in mice. The protection mediated by MSCs-Exo carrying miR-130b-3p could be rescued by elevating TGFBR1 expression. CONCLUSION miR-130b-3p delivery by MSCs-Exo confers protection on ALI in mice via the downregulation of TGFBR1.
Collapse
Affiliation(s)
- Xiaoxia Wang
- Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China.,The Laboratory of Perioperative Medicine Research Center, Guangxi Medical University Affiliated Tumor Hospital & Oncology Medical College, Nanning, Guangxi, China.,Department of Anesthesiology, Guangxi Medical University Affiliated Tumor Hospital & Oncology Medical College, Nanning, Guangxi, China.,Department of Anesthesiology, The Maternal and & Child Health Hospital, The Children's Hospital, The Obstetrics & Gynecology Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Jifeng Feng
- Department of Anesthesiology, The Maternal and & Child Health Hospital, The Children's Hospital, The Obstetrics & Gynecology Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Huijun Dai
- Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China.,The Laboratory of Perioperative Medicine Research Center, Guangxi Medical University Affiliated Tumor Hospital & Oncology Medical College, Nanning, Guangxi, China.,Department of Anesthesiology, Guangxi Medical University Affiliated Tumor Hospital & Oncology Medical College, Nanning, Guangxi, China.,Department of Anesthesiology, The Maternal and & Child Health Hospital, The Children's Hospital, The Obstetrics & Gynecology Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Jianla Mo
- Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China.,The Laboratory of Perioperative Medicine Research Center, Guangxi Medical University Affiliated Tumor Hospital & Oncology Medical College, Nanning, Guangxi, China.,Department of Anesthesiology, Guangxi Medical University Affiliated Tumor Hospital & Oncology Medical College, Nanning, Guangxi, China.,Department of Anesthesiology, The Maternal and & Child Health Hospital, The Children's Hospital, The Obstetrics & Gynecology Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Bijun Luo
- Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China.,The Laboratory of Perioperative Medicine Research Center, Guangxi Medical University Affiliated Tumor Hospital & Oncology Medical College, Nanning, Guangxi, China.,Department of Anesthesiology, Guangxi Medical University Affiliated Tumor Hospital & Oncology Medical College, Nanning, Guangxi, China.,Department of Anesthesiology, The Maternal and & Child Health Hospital, The Children's Hospital, The Obstetrics & Gynecology Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Cheng Luo
- Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
| | - Weikang Zhang
- The Affiliated Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Oncology and Basic Medicine, Chinese Academy of Sciences, Hangzhou City, Zhejiang Province, China
| | - Linghui Pan
- Key Laboratory for Basic Science and Prevention of Perioperative Organ Disfunction Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China.,The Laboratory of Perioperative Medicine Research Center, Guangxi Medical University Affiliated Tumor Hospital & Oncology Medical College, Nanning, Guangxi, China.,Department of Anesthesiology, Guangxi Medical University Affiliated Tumor Hospital & Oncology Medical College, Nanning, Guangxi, China
| |
Collapse
|
24
|
Wang Z, Yu T, Hou Y, Zhou W, Ding Y, Nie H. Mesenchymal Stem Cell Therapy for ALI/ARDS: Therapeutic Potential and Challenges. Curr Pharm Des 2022; 28:2234-2240. [PMID: 35796453 DOI: 10.2174/1381612828666220707104356] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 05/24/2022] [Indexed: 11/22/2022]
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is a serious clinical common disease, which may be caused by a variety of pathological factors and can induce a series of serious complications. There is still no specific and effective method for the treatment of ALI/ARDS. Mesenchymal stem cells (MSCs) have been one of the treatment methods for ALI, which can regulate related signal pathways such as PI3K/AKT, Wnt, and NF-κB to reduce inflammation. MSCs exist in a variety of tissues and have the ability of self-renewal and differentiation, which can be activated by specific substances or environments and home to the site of tissue damage, where they differentiate into new tissue cells and repair the damage. Both exosomes and cytokines involving the paracrine mechanism of MSCs have benefits on the treatment of ALI. Lung organoids produced by 3D culture technology can simulate the characteristics of the lung and help to research the pathophysiological process of ALI. This review summarizes the mechanisms by which MSCs treat ALI/ARDS and expects to use 3D models for future challenges in this field.
Collapse
Affiliation(s)
- Zhenxing Wang
- Department of Hematology and Breast Cancer, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Tong Yu
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yapeng Hou
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Wei Zhou
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yan Ding
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Hongguang Nie
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| |
Collapse
|
25
|
Khosravi-Farsani S, Zaminy A, Kazemi S, Hashemzadeh-Chaleshtori M. Mesenchymal stem cells versus their conditioned medium in the treatment of ischemia/reperfusion injury: Evaluation of efficacy and hepatic specific gene expression in mice. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:799-807. [PMID: 36033951 PMCID: PMC9392563 DOI: 10.22038/ijbms.2022.62642.13860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 06/26/2022] [Indexed: 11/05/2022]
Abstract
Objectives The mechanisms underlying the beneficial effects of MSCs on hepatic I/R injury are still poorly described, especially the changes in hepatocyte gene expression. In this study, the effect of bone marrow-derived mesenchymal stem cells (BMSCs) and adipose tissue-derived mesenchymal stem cells (AMSCs) and their conditioned medium on hepatocyte gene expression resulted by I/R shock were investigated. Materials and Methods Liver ischemia models were induced by clamping in experimental groups. Experimental groups received MSCs or conditioned medium treatments and the control group received Dulbecco's Modified Eagle Medium (DMEM). During 1, 24 hr, and 1 week after treatment, the serum levels of alanine aminotransferase (ALT), aspartate transaminase (AST) and lactate dehydrogenase (LDH) enzymes and tissue catalase activity (CAT) were measured. Gene expression of a number of hepatocyte-specific genes (Alb, Afp, and Ck8) and Icam-1 which is upregulated under inflammatory conditions were also evaluated in 5, 24 hr, and 1-week intervals after I/R insult. Results In this study, liver enzymes showed a much more shift in the control group than treated groups and it was more noticeable 5 hr post-treatment. Moreover, gene expression pattern of the control group underwent changes after I/R injury. However, treated groups gene expression analysis met a steady trend after I/R insult. Conclusion Our finding shows that stem cell treatment has better curative effects than conditioned medium. BMSCs, AMSCs or BMSC and AMSC-derived bioactive molecules injection have potential to be considered as a therapeutic approach for treating acute liver injury.
Collapse
Affiliation(s)
- Somayeh Khosravi-Farsani
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran, Department of Anatomical Sciences, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Arash Zaminy
- Burn and Regenerative Medicine Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Sedigheh Kazemi
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Morteza Hashemzadeh-Chaleshtori
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran,Corresponding author: Morteza Hashemzadeh-Chaleshtori. Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran. Tel: +98-38-33331471;
| |
Collapse
|
26
|
Ali NB, Abdull Razis AF, Ooi DJ, Chan KW, Ismail N, Foo JB. Theragnostic Applications of Mammal and Plant-Derived Extracellular Vesicles: Latest Findings, Current Technologies, and Prospects. Molecules 2022; 27:3941. [PMID: 35745063 PMCID: PMC9228370 DOI: 10.3390/molecules27123941] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/26/2022] [Accepted: 02/11/2022] [Indexed: 11/16/2022] Open
Abstract
The way cells communicate is not fully understood. However, it is well-known that extracellular vesicles (EVs) are involved. Researchers initially thought that EVs were used by cells to remove cellular waste. It is now clear that EVs function as signaling molecules released by cells to communicate with one another, carrying a cargo representing the mother cell. Furthermore, these EVs can be found in all biological fluids, making them the perfect non-invasive diagnostic tool, as their cargo causes functional changes in the cells upon receiving, unlike synthetic drug carriers. EVs last longer in circulation and instigate minor immune responses, making them the perfect drug carrier. This review sheds light on the latest development in EVs isolation, characterization and, application as therapeutic cargo, novel drug loading techniques, and diagnostic tools. We also address the advancement in plant-derived EVs, their characteristics, and applications; since plant-derived EVs only recently gained focus, we listed the latest findings. Although there is much more to learn about, EV is a wide field of research; what scientists have discovered so far is fascinating. This paper is suitable for those new to the field seeking to understand EVs and those already familiar with it but wanting to review the latest findings.
Collapse
Affiliation(s)
- Nada Basheir Ali
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia;
| | - Ahmad Faizal Abdull Razis
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia;
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia; (K.W.C.); (N.I.)
| | - Der Jiun Ooi
- Department of Oral Biology and Biomedical Sciences, Faculty of Dentistry, MAHSA University, Bandar Saujana Putra, Jenjarom 42610, Selangor, Malaysia
| | - Kim Wei Chan
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia; (K.W.C.); (N.I.)
| | - Norsharina Ismail
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia; (K.W.C.); (N.I.)
| | - Jhi Biau Foo
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya 47500, Selangor, Malaysia;
| |
Collapse
|
27
|
Song X, Xue Y, Fan S, Hao J, Deng R. Lipopolysaccharide-activated macrophages regulate the osteogenic differentiation of bone marrow mesenchymal stem cells through exosomes. PeerJ 2022; 10:e13442. [PMID: 35586136 PMCID: PMC9109694 DOI: 10.7717/peerj.13442] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 04/25/2022] [Indexed: 01/14/2023] Open
Abstract
Background Periodontal tissue regeneration is the ultimate goal of periodontitis treatment. Exosomes are nanoscale vesicles secreted by cells that participate in and regulate the physiological activities between cells. However, the relationship between inflammatory macrophage-derived exosomes and osteoblast differentiation in periodontitis has not been thoroughly reported. Here, we attempt to explore the role of inflammatory macrophage-derived exosomes in crosstalk with osteoblasts. Methods Porphyromonas gingivalis lipopolysaccharide was used to stimulate macrophages and inflate their inflammatory cellular state. Exosomes were extracted from inflammatory macrophages using supercentrifugation, and their characteristics were detected by transmission electron microscopy, particle size analysis, and Western blotting. Exosome uptake bybone marrow mesenchymal stem cells (BMSCs) was observed by fluorescence microscopy. The effects of exosomes on the BMSC inflammatory response and on osteogenic differentiation were detected by quantitative polymerase chain reaction and Western blot analysis. Alkaline phosphatase activity was tested for verification. Results We successfully extracted and identified inflammatory macrophage-derived exosomes and observed that BMSCs successfully took up exosomes. Inflammatory macrophage-derived exosomes upregulated the expression levels of the inflammatory factors interleukin-6 and tumour necrosis factor-alpha in BMSCs and mediated inflammatory stimulation. Additionally, they inhibited the transcription levels of the osteogenic genes alkaline phosphatase, type I collagen, and Runt-related transcription factor 2 as well as the alkaline phosphatase activity, while the use of the exosome inhibitor GW4869 attenuated this effect. Conclusion Our study shows that macrophages in periodontitis can mediate inflammatory stimulation and inhibit the osteogenic differentiation of bone marrow mesenchymal stem cells through the exosome pathway. Interference with exosome secretion is likely to be a promising method for bone tissue regeneration in inflammatory states.
Collapse
Affiliation(s)
- Xiao Song
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China,Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Yiwen Xue
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China,Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Siyu Fan
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China,Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Jing Hao
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Runzhi Deng
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| |
Collapse
|
28
|
Hua Y, Han A, Yu T, Hou Y, Ding Y, Nie H. Small Extracellular Vesicles Containing miR-34c Derived from Bone Marrow Mesenchymal Stem Cells Regulates Epithelial Sodium Channel via Targeting MARCKS. Int J Mol Sci 2022; 23:ijms23095196. [PMID: 35563590 PMCID: PMC9101277 DOI: 10.3390/ijms23095196] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/29/2022] [Accepted: 05/04/2022] [Indexed: 12/18/2022] Open
Abstract
Epithelial sodium channel (ENaC) is a pivotal regulator of alveolar fluid clearance in the airway epithelium and plays a key role in the treatment of acute lung injury (ALI), which is mainly composed of the three homologous subunits (α, β and γ). The mechanisms of microRNAs in small extracellular vesicles (sEVs) derived from mesenchymal stem cell (MSC-sEVs) on the regulation of lung ion transport are seldom reported. In this study, we aimed at investigating whether miR-34c had an effect on ENaC dysfunction induced by lipopolysaccharide and explored the underlying mechanism in this process. Primarily, the effect of miR-34c on lung edema and histopathology changes in an ALI mouse model was investigated. Then the uptake of PKH26-labeled sEVs was observed in recipient cells, and we observed that the overexpression of miR-34c in MSC-sEVs could upregulate the LPS-inhibited γ-ENaC expression. The dual luciferase reporter gene assay demonstrated that myristoylated alanine-rich C kinase substrate (MARCKS) was one of target genes of miR-34c, the protein expression of which was negatively correlated with miR-34c. Subsequently, either upregulating miR-34c or knocking down MARCKS could increase the protein expression of phospho-phosphatidylinositol 3-kinase (p-PI3K) and phospho-protein kinase B (p-AKT), implying a downstream regulation pathway was involved. All of the above suggest that miR-34c in MSC-sEVs can attenuate edematous lung injury via enhancing γ-ENaC expression, at least partially, through targeting MARCKS and activating the PI3K/AKT signaling pathway subsequently.
Collapse
|
29
|
Mesenchymal Stem Cell-Based Therapy as a New Approach for the Treatment of Systemic Sclerosis. Clin Rev Allergy Immunol 2022; 64:284-320. [PMID: 35031958 DOI: 10.1007/s12016-021-08892-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2021] [Indexed: 12/13/2022]
Abstract
Systemic sclerosis (SSc) is an intractable autoimmune disease with unmet medical needs. Conventional immunosuppressive therapies have modest efficacy and obvious side effects. Targeted therapies with small molecules and antibodies remain under investigation in small pilot studies. The major breakthrough was the development of autologous haematopoietic stem cell transplantation (AHSCT) to treat refractory SSc with rapidly progressive internal organ involvement. However, AHSCT is contraindicated in patients with advanced visceral involvement. Mesenchymal stem cells (MSCs) which are characterized by immunosuppressive, antifibrotic and proangiogenic capabilities may be a promising alternative option for the treatment of SSc. Multiple preclinical and clinical studies on the use of MSCs to treat SSc are underway. However, there are several unresolved limitations and safety concerns of MSC transplantation, such as immune rejections and risks of tumour formation, respectively. Since the major therapeutic potential of MSCs has been ascribed to their paracrine signalling, the use of MSC-derived extracellular vesicles (EVs)/secretomes/exosomes as a "cell-free" therapy might be an alternative option to circumvent the limitations of MSC-based therapies. In the present review, we overview the current knowledge regarding the therapeutic efficacy of MSCs in SSc, focusing on progresses reported in preclinical and clinical studies using MSCs, as well as challenges and future directions of MSC transplantation as a treatment option for patients with SSc.
Collapse
|
30
|
Alsaadi N, Srinivasan AJ, Seshadri A, Shiel M, Neal MD, Scott MJ. The emerging therapeutic potential of extracellular vesicles in trauma. J Leukoc Biol 2022; 111:93-111. [PMID: 34533241 PMCID: PMC9169334 DOI: 10.1002/jlb.3mir0621-298r] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Traumatic injury is a major cause of morbidity and mortality worldwide, despite significant advances in treatments. Most deaths occur either very early, through massive head trauma/CNS injury or exsanguination (despite advances in transfusion medicine), or later after injury often through multiple organ failure and secondary infection. Extracellular vesicles (EVs) are known to increase in the circulation after trauma and have been used to limited extent as diagnostic and prognostic markers. More intriguingly, EVs are now being investigated as both causes of pathologies post trauma, such as trauma-induced coagulopathy, and as potential treatments. In this review, we highlight what is currently known about the role and effects of EVs in various aspects of trauma, as well as exploring current literature from investigators who have begun to use EVs therapeutically to alter the physiology and pathology of traumatic insults. The potential effectiveness of using EVs therapeutically in trauma is supported by a large number of experimental studies, but there is still some way to go before we understand the complex effects of EVs in what is already a complex disease process.
Collapse
Affiliation(s)
- Nijmeh Alsaadi
- Division of General and Trauma Surgery, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Amudan J Srinivasan
- Division of General and Trauma Surgery, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Anupamaa Seshadri
- Division of General and Trauma Surgery, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Matthew Shiel
- Division of Hematology-Oncology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Matthew D Neal
- Division of General and Trauma Surgery, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Pittsburgh Trauma Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Melanie J Scott
- Division of General and Trauma Surgery, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Pittsburgh Trauma Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
31
|
Tan TT, Toh WS, Lai RC, Lim SK. Practical considerations in transforming MSC therapy for neurological diseases from cell to EV. Exp Neurol 2021; 349:113953. [PMID: 34921846 DOI: 10.1016/j.expneurol.2021.113953] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/19/2021] [Accepted: 12/13/2021] [Indexed: 12/11/2022]
Abstract
Cell-based therapy using Mesenchymal Stromal Cell (MSC) has generally been efficacious in treating a myriad of diseases in animal models and clinical trials. The rationale for MSC therapy was predicated on the potential of MSC to differentiate and form new replacement cells in the diseased tissue. However, pre-clinical animal and clinical data were more consistent with a secretion- and not a differentiation-based rationale. Analysis of MSC secretion led to the identification of small extracellular vesicles (sEVs) as therapeutically active, secretory agents. MSC-sEVs are defined as bi-lipid membrane vesicles of 50-200 nm in diameter that are secreted by MSCs. They reportedly exert similar therapeutic efficacy as MSCs in many diseases including neurological diseases. MSC-sEVs being small and non-living are intrinsically safer than living MSCs. Manufacturing of MSC-sEVs may also be less complex. Nevertheless, realising the therapeutic potential of MSC-sEVs will require exacting scientific rigor and robustness, as well as compliance to regulatory oversight. This review summarises the scientific rationale for the transition of MSC therapy from a cell- to an EV-based therapy and discusses critical scientific issues in the development of MSC-sEVs therapy.
Collapse
Affiliation(s)
- Thong Teck Tan
- Institute of Molecular and Cellular Biology, A*STAR, 8A Biomedical Grove, Singapore 138648, Singapore
| | - Wei Seong Toh
- Faculty of Dentistry, National University of Singapore, Singapore; Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Ruenn Chai Lai
- Institute of Molecular and Cellular Biology, A*STAR, 8A Biomedical Grove, Singapore 138648, Singapore
| | - Sai Kiang Lim
- Institute of Molecular and Cellular Biology, A*STAR, 8A Biomedical Grove, Singapore 138648, Singapore; Department of Surgery, YLL School of Medicine, National University of Singapore (NUS) Lower Kent Ridge Road, Singapore 119074, Singapore.
| |
Collapse
|
32
|
Willis GR, Reis M, Gheinani AH, Fernandez-Gonzalez A, Taglauer ES, Yeung V, Liu X, Ericsson M, Haas E, Mitsialis SA, Kourembanas S. Extracellular Vesicles Protect the Neonatal Lung from Hyperoxic Injury through the Epigenetic and Transcriptomic Reprogramming of Myeloid Cells. Am J Respir Crit Care Med 2021; 204:1418-1432. [PMID: 34699335 PMCID: PMC8865710 DOI: 10.1164/rccm.202102-0329oc] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 10/18/2021] [Indexed: 11/16/2022] Open
Abstract
Rationale: Mesenchymal stem/stromal cell (MSC)-small extracellular vesicle (MEx) treatment has shown promise in experimental models of neonatal lung injury. The molecular mechanisms by which MEx afford beneficial effects remain incompletely understood. Objectives: To investigate the therapeutic mechanism of action through assessment of MEx biodistribution and impact on immune cell phenotypic heterogeneity. Methods: MEx were isolated from the conditioned medium of human umbilical cord Wharton's jelly-derived MSCs. Newborn mice were exposed to hyperoxia (HYRX, 75% O2) from birth and returned to room air at Postnatal Day 14 (PN14). Mice received either a bolus intravenous MEx dose at PN4 or bone marrow-derived myeloid cells (BMDMy) pretreated with MEx. Animals were killed at PN4, PN7, PN14, or PN28 to characterize MEx biodistribution or for assessment of pulmonary parameters. The therapeutic role of MEx-educated BMDMy was determined in vitro and in vivo. Measurements and Main Results: MEx therapy ameliorated core histological features of HYRX-induced neonatal lung injury. Biodistribution and mass cytometry studies demonstrated that MEx localize in the lung and interact with myeloid cells. MEx restored the apportion of alveolar macrophages in the HYRX-injured lung and concomitantly suppressed inflammatory cytokine production. In vitro and ex vivo studies revealed that MEx promoted an immunosuppressive BMDMy phenotype. Functional assays demonstrated that the immunosuppressive actions of BMDMy are driven by phenotypically and epigenetically reprogrammed monocytes. Adoptive transfer of MEx-educated BMDMy, but not naive BMDMy, restored alveolar architecture, blunted fibrosis and pulmonary vascular remodeling, and improved exercise capacity. Conclusions: MEx ameliorate hyperoxia-induced neonatal lung injury though epigenetic and phenotypic reprogramming of myeloid cells.
Collapse
Affiliation(s)
- Gareth R. Willis
- Division of Newborn Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Monica Reis
- Division of Newborn Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Ali Hashemi Gheinani
- Department of Urology, Boston Children’s Hospital, Boston, Massachusetts
- Department of Surgery, Harvard Medical School, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Angeles Fernandez-Gonzalez
- Division of Newborn Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Elizabeth S. Taglauer
- Division of Newborn Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Vincent Yeung
- Division of Newborn Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Xianlan Liu
- Division of Newborn Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts
| | - Maria Ericsson
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts; and
| | - Eric Haas
- Mass Cytometry Core, Dana Farber Cancer Institute, Boston, Massachusetts
| | - S. Alex Mitsialis
- Division of Newborn Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| | - Stella Kourembanas
- Division of Newborn Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, Massachusetts
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
33
|
Esquivel-Ruiz S, González-Rodríguez P, Lorente JA, Pérez-Vizcaíno F, Herrero R, Moreno L. Extracellular Vesicles and Alveolar Epithelial-Capillary Barrier Disruption in Acute Respiratory Distress Syndrome: Pathophysiological Role and Therapeutic Potential. Front Physiol 2021; 12:752287. [PMID: 34887773 PMCID: PMC8650589 DOI: 10.3389/fphys.2021.752287] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/27/2021] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EVs) mediate intercellular communication by transferring genetic material, proteins and organelles between different cells types in both health and disease. Recent evidence suggests that these vesicles, more than simply diagnostic markers, are key mediators of the pathophysiology of acute respiratory distress syndrome (ARDS) and other lung diseases. In this review, we will discuss the contribution of EVs released by pulmonary structural cells (alveolar epithelial and endothelial cells) and immune cells in these diseases, with particular attention to their ability to modulate inflammation and alveolar-capillary barrier disruption, a hallmark of ARDS. EVs also offer a unique opportunity to develop new therapeutics for the treatment of ARDS. Evidences supporting the ability of stem cell-derived EVs to attenuate the lung injury and ongoing strategies to improve their therapeutic potential are also discussed.
Collapse
Affiliation(s)
- Sergio Esquivel-Ruiz
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Paloma González-Rodríguez
- Ciber de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Department of Critical Care, Hospital Universitario de Getafe, Madrid, Spain
| | - José A Lorente
- Ciber de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Department of Critical Care, Hospital Universitario de Getafe, Madrid, Spain.,Clinical Section, School of Medicine, European University of Madrid, Madrid, Spain
| | - Francisco Pérez-Vizcaíno
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Raquel Herrero
- Ciber de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Department of Critical Care, Hospital Universitario de Getafe, Madrid, Spain
| | - Laura Moreno
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| |
Collapse
|
34
|
Mao GC, Gong CC, Wang Z, Sun MX, Pei ZP, Meng WQ, Cen JF, He XW, Lu Y, Xu QQ, Xiao K. BMSC-derived exosomes ameliorate sulfur mustard-induced acute lung injury by regulating the GPRC5A-YAP axis. Acta Pharmacol Sin 2021; 42:2082-2093. [PMID: 33654219 PMCID: PMC8633287 DOI: 10.1038/s41401-021-00625-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 02/09/2021] [Indexed: 12/12/2022] Open
Abstract
Sulfur mustard (SM) is a highly toxic chemical warfare agent that causes acute lung injury (ALI) and/or acute respiratory distress syndrome (ARDS). There are no effective therapeutic treatments or antidotes available currently to counteract its toxic effects. Our previous study shows that bone marrow-derived mesenchymal stromal cells (BMSCs) could exert therapeutic effects against SM-induced lung injury. In this study, we explored the therapeutic potential of BMSC-derived exosomes (BMSC-Exs) against ALI and the underlying mechanisms. ALI was induced in mice by injection of SM (30 mg/kg, sc) at their medial and dorsal surfaces. BMSC-Exs (20 μg/kg in 200 μL PBS, iv) were injected for a 5-day period after SM exposure. We showed that BMSC-Exs administration caused a protective effect against pulmonary edema. Using a lung epithelial cell barrier model, BMSC-Exs (10, 20, 40 μg) dose-dependently inhibited SM-induced cell apoptosis and promoted the recovery of epithelial barrier function by facilitating the expression and relocalization of junction proteins (E-cadherin, claudin-1, occludin, and ZO-1). We further demonstrated that BMSC-Exs protected against apoptosis and promoted the restoration of barrier function against SM through upregulating G protein-coupled receptor family C group 5 type A (GPRC5A), a retinoic acid target gene predominately expressed in the epithelial cells of the lung. Knockdown of GPRC5A reduced the antiapoptotic and barrier regeneration abilities of BMSC-Exs and diminished their therapeutic effects in vitro and in vivo. BMSC-Exs-caused upregulation of GPRC5A promoted the expression of Bcl-2 and junction proteins via regulating the YAP pathway. In summary, BMSC-Exs treatment exerts protective effects against SM-induced ALI by promoting alveolar epithelial barrier repair and may be an alternative approach to stem cell-based therapy.
Collapse
Affiliation(s)
- Guan-Chao Mao
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Chu-Chu Gong
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Zhen Wang
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
- Department of Preventive Medicinne, School of Medicine, Hunan Normal University, Changsha, China
| | - Ming-Xue Sun
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Zhi-Peng Pei
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Wen-Qi Meng
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Jin-Feng Cen
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Xiao-Wen He
- Origincell Technology Group Co., Ltd., Shanghai, 201203, China
| | - Ying Lu
- Department of Pharmaceutical Science, School of Pharmacy, Naval Medical University, Shanghai, 200433, China.
| | - Qing-Qiang Xu
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China.
| | - Kai Xiao
- Lab of Toxicology and Pharmacology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
35
|
Protective effects of low-molecular-weight components of adipose stem cell-derived conditioned medium on dry eye syndrome in mice. Sci Rep 2021; 11:21874. [PMID: 34750552 PMCID: PMC8575953 DOI: 10.1038/s41598-021-01503-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 10/29/2021] [Indexed: 11/08/2022] Open
Abstract
The present study demonstrated the protective effects of low-molecular-weight adipose-derived stem cell-conditioned medium (LADSC-CM) in a mouse model of dry eye syndrome. Mice subjected to desiccating stress and benzalkonium chloride had decreased tear secretion, impaired corneal epithelial tight junction with microvilli, and decreased conjunctival goblet cells. Topical application of adipose-derived stem cell-conditioned medium (ADSC-CM) stimulated lacrimal tear secretion, preserved tight junction and microvilli of the corneal epithelium, and increased the density of goblet cells and MUC16 expression in the conjunctiva. The low-molecular-weight fractions (< 10 kDa and < 3 kDa) of ADSC-CM (LADSC-CM) provided better protections than the > 10 kDa or > 3 kDa fractions of ADSC-CM. In the in vitro study, desiccation for 10 min or hyperosmolarity (490 osmols) for 24 h caused decreased viability of human corneal epithelial cells, which were reversed by LADSC-CM. The active ingredients in the LADSC-CM were lipophobic and stable after heating and lyophilization. Our study demonstrated that LADSC-CM had beneficial effects on experimental dry eye. It is worthy of further exploration for the active ingredient(s) and the mechanism.
Collapse
|
36
|
Zhang X, Sugita S, Liu A, Naito Y, Hwang W, Qiu H, Sakamoto A, Sawa T, Matthay MA, Lee JW. Therapeutic effects of high molecular weight hyaluronic acid in severe Pseudomonas aeruginosa pneumonia in ex vivo perfused human lungs. Am J Physiol Lung Cell Mol Physiol 2021; 321:L827-L836. [PMID: 34524905 DOI: 10.1152/ajplung.00626.2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We previously reported that extracellular vesicles (EVs) released during Escherichia coli (E. coli) bacterial pneumonia were inflammatory, and administration of high molecular weight hyaluronic acid (HMW HA) suppressed several indices of acute lung injury (ALI) from E. coli pneumonia by binding to these inflammatory EVs. The current study was undertaken to study the therapeutic effects of HMW HA in ex vivo perfused human lungs injured with Pseudomonas aeruginosa (PA)103 bacterial pneumonia. For lungs with baseline alveolar fluid clearance (AFC) <10%/h, HMW HA 1 or 2 mg was injected intravenously after 1 h (n = 4-9), and EVs released during PA pneumonia were collected from the perfusate over 6 h. For lungs with baseline AFC > 10%/h, HMW HA 2 mg was injected intravenously after 1 h (n = 6). In vitro experiments were conducted to evaluate the effects of HA on inflammation and bacterial phagocytosis. For lungs with AFC < 10%/h, administration of HMW HA intravenously significantly restored AFC and numerically decreased protein permeability and alveolar inflammation from PA103 pneumonia but had no effect on bacterial counts at 6 h. However, HMW HA improved bacterial phagocytosis by human monocytes and neutrophils and suppressed the inflammatory properties of EVs released during pneumonia on monocytes. For lungs with AFC > 10%/h, administration of HMW HA intravenously improved AFC from PA103 pneumonia but had no significant effects on protein permeability, inflammation, or bacterial counts. In the presence of impaired alveolar epithelial transport capacity, administration of HMW HA improved the resolution of pulmonary edema from Pseudomonas PA103 bacterial pneumonia.
Collapse
Affiliation(s)
- Xiwen Zhang
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China.,Departments of Anesthesiology, Medicine, and Cardiovascular Research Institute, University of California, San Francisco, California
| | - Shinji Sugita
- Department of Anesthesiology, Nippon Medical School, Tokyo, Japan.,Departments of Anesthesiology, Medicine, and Cardiovascular Research Institute, University of California, San Francisco, California
| | - Airan Liu
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Yoshifumi Naito
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Wonjung Hwang
- Departments of Anesthesiology, Medicine, and Cardiovascular Research Institute, University of California, San Francisco, California
| | - Haibo Qiu
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | | | - Teiji Sawa
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Michael A Matthay
- Departments of Anesthesiology, Medicine, and Cardiovascular Research Institute, University of California, San Francisco, California
| | - Jae-Woo Lee
- Departments of Anesthesiology, Medicine, and Cardiovascular Research Institute, University of California, San Francisco, California
| |
Collapse
|
37
|
Gu G, Lv X, Liu G, Zeng R, Li S, Chen L, Liang Z, Wang H, Lu F, Zhan L, Lv X. Tnfaip6 Secreted by Bone Marrow-Derived Mesenchymal Stem Cells Attenuates TNBS-Induced Colitis by Modulating Follicular Helper T Cells and Follicular Regulatory T Cells Balance in Mice. Front Pharmacol 2021; 12:734040. [PMID: 34707499 PMCID: PMC8542666 DOI: 10.3389/fphar.2021.734040] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/31/2021] [Indexed: 12/16/2022] Open
Abstract
Objective: To investigate the immunological mechanism of bone marrow-derived mesenchymal stem cells (BM-MSCs) in inflammatory bowel disease (IBD). Methods: Mice with 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced colitis were intraperitoneally injected with phosphate-buffered saline, BM-MSCs, BM-MSCs with tumor necrosis factor-induced protein 6 (Tnfaip6) knockdown mediated by RNA interference recombinant adenovirus, and BM-MSCs-infected with control adenovirus or recombinant mouse Tnfaip6. The disease activity index, weight loss, and histological scores were recorded. Serum levels of Tnfaip6 and pro- and anti-inflammatory cytokines, including interleukin (IL)-21, tumor necrosis factor-alpha (TNF-α), IL-10 were measured by enzyme-linked immunosorbent assay. The relative expression levels of these cytokines, B-cell lymphoma 6 (BCL-6) and fork-like transcription factor p3 (Foxp3) in the colon were determined by real-time quantitative PCR (RT-qPCR). BCL-6 and Foxp3 are the master regulators of follicular helper T cells (Tfh) and follicular regulatory T cells (Tfr), respectively. The infiltration of Tfh and Tfr in mesenteric lymph nodes (MLNs) and spleens was analyzed by flow cytometry. Results: Compared to the normal control group, the expression levels of BCL-6 and IL-21 in the colon, Tfh infiltration, and ratios of Tfh/Tfr in the MLNs and spleen, and the serum concentrations of IL-21 and TNF-α increased significantly in the colitis model group (p < 0.05). Intraperitoneal injection of BM-MSCs or Tnfaip6 ameliorated weight loss and clinical and histological severity of colitis, downregulated the expression of BCL-6, IL-21, and TNF-α, upregulated the expression of Foxp3, IL-10, and Tnfaip6 (p < 0.05), increased Tfr and reduced the infiltration of Tfh in the MLNs and spleen, and downregulated the Tfh/Tfr ratio (p < 0.05). On the other hand, BM-MSCs lost the therapeutic effect and immune regulatory functions on Tfh and Tfr after Tnfaip6 knockdown. Conclusion: Tfh increase in the inflamed colon, Tfh decrease and Tfr increase during the colitis remission phase, and the imbalance of the Tfh/Tfr ratio is closely related to the progression of IBD. Tnfaip6 secreted by BM-MSCs alleviates IBD by inhibiting Tfh differentiation, promoting Tfr differentiation, and improving the imbalance of Tfh/Tfr in mice.
Collapse
Affiliation(s)
- Guangli Gu
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiaodan Lv
- Department of Clinical Experimental Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Gengfeng Liu
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ruizhi Zeng
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shiquan Li
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lan Chen
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhaoliang Liang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Huiqin Wang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Fei Lu
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lingling Zhan
- Department of Clinical Experimental Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiaoping Lv
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
38
|
Qi LL, Fan ZY, Mao HG, Wang JB. The Therapeutic Efficacy of Adipose Tissue-Derived Mesenchymal Stem Cell Conditioned Medium on Experimental Colitis Was Improved by the Serum From Colitis Rats. Front Bioeng Biotechnol 2021; 9:694908. [PMID: 34604183 PMCID: PMC8484792 DOI: 10.3389/fbioe.2021.694908] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 09/06/2021] [Indexed: 01/07/2023] Open
Abstract
Adipose derived mesenchymal stem cells (AD-MSCs) have shown therapeutic potential in treatments of inflammatory bowel disease (IBD). Due to the harsh host environment and poor survival of the cells, controversy concerning the homing, proliferation and differentiation of MSCs in lesion tissue still remains. It has been reported that conditioned media from MSCs could improve the colitis, whereas the therapeutic efficiency could be significantly elevated by the stimulation of pro-cytokines. In this study, we pre-treated the adipose derived MSCs with the serum from colitis rats and then the activated conditioned media (CM-AcMSC) were collected. To compare the therapeutic effects of CM-MSC and CM-AcMSC on IBD, we constructed dextran sodium sulphate (DSS)-induced colitis rat models. The colitis was induced in rats by administrating 5% DSS in drinking water for 10 days, and the disease symptoms were recorded daily. The colon histopathological changes were observed by different staining methods (H&E and PAS). The expression levels of MUC2 and tight junctions (TJs) were determined by RT-qPCR. The levels of inflammatory cytokines were analyzed by ELISA and western blot analysis. Our findings suggested that CM-AcMSC was more effective in ameliorating the clinical features and histological damage scores. Treatment with CM-AcMSC significantly increased the expression of MUC2 and TJs and suppressed the production of pro-inflammatory cytokines in colonic tissues of colitis rats. The inhibitory effects of CM-AcMSC on inflammatory responses of colitis rats were mediated by NF-κB signaling pathway. These results suggested that pre-activation of MSCs with serum from colitis rats could promote the production of paracrine factors and improve the therapeutic effects of conditioned medium on colitis rats.
Collapse
Affiliation(s)
- Li-Li Qi
- School of Biological and Chemical Engineering, Ningbo Tech University, Ningbo, China
| | - Zhe-Yu Fan
- School of Biological and Chemical Engineering, Ningbo Tech University, Ningbo, China
| | - Hai-Guang Mao
- School of Biological and Chemical Engineering, Ningbo Tech University, Ningbo, China
| | - Jin-Bo Wang
- School of Biological and Chemical Engineering, Ningbo Tech University, Ningbo, China
| |
Collapse
|
39
|
Tieu A, Hu K, Gnyra C, Montroy J, Fergusson DA, Allan DS, Stewart DJ, Thébaud B, Lalu MM. Mesenchymal stromal cell extracellular vesicles as therapy for acute and chronic respiratory diseases: A meta-analysis. J Extracell Vesicles 2021; 10:e12141. [PMID: 34596349 PMCID: PMC8485337 DOI: 10.1002/jev2.12141] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/30/2021] [Accepted: 08/23/2021] [Indexed: 12/15/2022] Open
Abstract
Preclinical studies suggest mesenchymal stromal cell extracellular vesicles (MSC-EVs) reduce inflammation and improve organ function in lung diseases; however, an objective analysis of all available data is needed prior to translation. Using rigorous meta-research methods, we determined the effectiveness of MSC-EVs for preclinical respiratory diseases and identified experimental conditions that may further refine this therapy. A systematic search of MEDLINE/Embase identified 1167 records. After screening, 52 articles were included for data extraction and evaluated for risk of bias and quality of reporting in study design. A random effects meta-analysis was conducted for acute lung injury (ALI; N = 23), bronchopulmonary dysplasia (BPD; N = 8) and pulmonary arterial hypertension (PAH; N = 7). Subgroup analyses identified EV methods/characteristics that may be associated with improved efficacy. Data is presented as standardized mean differences (SMD) or risk ratios (RR) with 95% confidence intervals (CI). For ALI, MSC-EVs markedly reduced lung injury (SMD -4.33, CI -5.73 to -2.92), vascular permeability (SMD -2.43, CI -3.05 to -1.82), and mortality (RR 0.39, CI 0.22 to 0.68). Small EVs were more consistently effective than large EVs whereas no differences were observed between tissue sources, immunocompatibility or isolation techniques. For BPD, alveolarization was improved by MSC-EVs (SMD -1.45, CI -2.08 to -0.82) with small EVs more consistently beneficial then small/large EVs. In PAH, right ventricular systolic pressure (SMD -4.16, CI -5.68 to -2.64) and hypertrophy (SMD -2.80, CI -3.68 to -1.91) were significantly attenuated by EVs. In BPD and PAH, EVs isolated by ultracentrifugation demonstrated therapeutic benefit whereas tangential flow filtration (N = 2) displayed minimal efficacy. Lastly, risk of bias and quality of reporting for experimental design were consistently unclear across all studies. Our findings demonstrate clear potential of MSC-EVs to be developed as therapy for acute and chronic lung diseases. However, greater transparency in research design and direct comparisons of isolation technique and EV subtypes are needed to generate robust evidence to guide clinical translation. Protocol Registration: PROSPERO CRD42020145334.
Collapse
Affiliation(s)
- Alvin Tieu
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaOntarioCanada
- Clinical Epidemiology ProgramBLUEPRINT Translational Research Group, Ottawa Hospital Research InstituteOttawaOntarioCanada
- Regenerative Medicine ProgramOttawa Hospital Research InstituteOttawaOntarioCanada
| | - Kevin Hu
- Clinical Epidemiology ProgramBLUEPRINT Translational Research Group, Ottawa Hospital Research InstituteOttawaOntarioCanada
| | - Catherine Gnyra
- Clinical Epidemiology ProgramBLUEPRINT Translational Research Group, Ottawa Hospital Research InstituteOttawaOntarioCanada
| | - Joshua Montroy
- Clinical Epidemiology ProgramBLUEPRINT Translational Research Group, Ottawa Hospital Research InstituteOttawaOntarioCanada
| | - Dean A. Fergusson
- Clinical Epidemiology ProgramBLUEPRINT Translational Research Group, Ottawa Hospital Research InstituteOttawaOntarioCanada
- Department of MedicineThe Ottawa HospitalOttawaOntarioCanada
| | - David S. Allan
- Clinical Epidemiology ProgramBLUEPRINT Translational Research Group, Ottawa Hospital Research InstituteOttawaOntarioCanada
- Regenerative Medicine ProgramOttawa Hospital Research InstituteOttawaOntarioCanada
- Department of MedicineThe Ottawa HospitalOttawaOntarioCanada
| | - Duncan J. Stewart
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaOntarioCanada
- Regenerative Medicine ProgramOttawa Hospital Research InstituteOttawaOntarioCanada
- Department of MedicineThe Ottawa HospitalOttawaOntarioCanada
| | - Bernard Thébaud
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaOntarioCanada
- Regenerative Medicine ProgramOttawa Hospital Research InstituteOttawaOntarioCanada
- Division of NeonatologyDepartment of PediatricsChildren's Hospital of Eastern OntarioOttawaOntarioCanada
| | - Manoj M. Lalu
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaOntarioCanada
- Clinical Epidemiology ProgramBLUEPRINT Translational Research Group, Ottawa Hospital Research InstituteOttawaOntarioCanada
- Regenerative Medicine ProgramOttawa Hospital Research InstituteOttawaOntarioCanada
- Departments of Anesthesiology and Pain Medicine, The Ottawa HospitalOttawaOntarioCanada
| |
Collapse
|
40
|
Qiao Q, Liu X, Yang T, Cui K, Kong L, Yang C, Zhang Z. Nanomedicine for acute respiratory distress syndrome: The latest application, targeting strategy, and rational design. Acta Pharm Sin B 2021; 11:3060-3091. [PMID: 33977080 PMCID: PMC8102084 DOI: 10.1016/j.apsb.2021.04.023] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 03/22/2021] [Accepted: 04/06/2021] [Indexed: 01/08/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is characterized by the severe inflammation and destruction of the lung air-blood barrier, leading to irreversible and substantial respiratory function damage. Patients with coronavirus disease 2019 (COVID-19) have been encountered with a high risk of ARDS, underscoring the urgency for exploiting effective therapy. However, proper medications for ARDS are still lacking due to poor pharmacokinetics, non-specific side effects, inability to surmount pulmonary barrier, and inadequate management of heterogeneity. The increased lung permeability in the pathological environment of ARDS may contribute to nanoparticle-mediated passive targeting delivery. Nanomedicine has demonstrated unique advantages in solving the dilemma of ARDS drug therapy, which can address the shortcomings and limitations of traditional anti-inflammatory or antioxidant drug treatment. Through passive, active, or physicochemical targeting, nanocarriers can interact with lung epithelium/endothelium and inflammatory cells to reverse abnormal changes and restore homeostasis of the pulmonary environment, thereby showing good therapeutic activity and reduced toxicity. This article reviews the latest applications of nanomedicine in pre-clinical ARDS therapy, highlights the strategies for targeted treatment of lung inflammation, presents the innovative drug delivery systems, and provides inspiration for strengthening the therapeutic effect of nanomedicine-based treatment.
Collapse
Key Words
- ACE2, angiotensin-converting enzyme 2
- AEC II, alveolar type II epithelial cells
- AM, alveolar macrophages
- ARDS, acute respiratory distress syndrome
- Acute lung injury
- Acute respiratory distress syndrome
- Anti-inflammatory therapy
- BALF, bronchoalveolar lavage fluid
- BSA, bovine serum albumin
- CD, cyclodextrin
- CLP, cecal ligation and perforation
- COVID-19
- COVID-19, coronavirus disease 2019
- DOPE, phosphatidylethanolamine
- DOTAP, 1-diolefin-3-trimethylaminopropane
- DOX, doxorubicin
- DPPC, dipalmitoylphosphatidylcholine
- Drug delivery
- ECM, extracellular matrix
- ELVIS, extravasation through leaky vasculature and subsequent inflammatory cell-mediated sequestration
- EPCs, endothelial progenitor cells
- EPR, enhanced permeability and retention
- EVs, extracellular vesicles
- EphA2, ephrin type-A receptor 2
- Esbp, E-selectin-binding peptide
- FcgR, Fcγ receptor
- GNP, peptide-gold nanoparticle
- H2O2, hydrogen peroxide
- HO-1, heme oxygenase-1
- ICAM-1, intercellular adhesion molecule-1
- IKK, IκB kinase
- IL, interleukin
- LPS, lipopolysaccharide
- MERS, Middle East respiratory syndrome
- MPMVECs, mouse pulmonary microvascular endothelial cells
- MPO, myeloperoxidase
- MSC, mesenchymal stem cells
- NAC, N-acetylcysteine
- NE, neutrophil elastase
- NETs, neutrophil extracellular traps
- NF-κB, nuclear factor-κB
- Nanomedicine
- PC, phosphatidylcholine
- PCB, poly(carboxybetaine)
- PDA, polydopamine
- PDE4, phosphodiesterase 4
- PECAM-1, platelet-endothelial cell adhesion molecule
- PEG, poly(ethylene glycol)
- PEI, polyetherimide
- PEVs, platelet-derived extracellular vesicles
- PLGA, poly(lactic-co-glycolic acid)
- PS-PEG, poly(styrene-b-ethylene glycol)
- Pathophysiologic feature
- RBC, red blood cells
- RBD, receptor-binding domains
- ROS, reactive oxygen species
- S1PLyase, sphingosine-1-phosphate lyase
- SARS, severe acute respiratory syndrome
- SARS-CoV-2, severe acute respiratory syndrome coronavirus 2
- SDC1, syndecan-1
- SORT, selective organ targeting
- SP, surfactant protein
- Se, selenium
- Siglec, sialic acid-binding immunoglobulin-like lectin
- TLR, toll-like receptor
- TNF-α, tumor necrosis factor-α
- TPP, triphenylphosphonium cation
- Targeting strategy
- YSA, YSAYPDSVPMMS
- cRGD, cyclic arginine glycine-d-aspartic acid
- iNOS, inducible nitric oxide synthase
- rSPANb, anti-rat SP-A nanobody
- scFv, single chain variable fragments
Collapse
Affiliation(s)
- Qi Qiao
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiong Liu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ting Yang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Kexin Cui
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Li Kong
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Conglian Yang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhiping Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Engineering Research Center for Novel Drug Delivery System, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
41
|
Liu JH, Li C, Cao L, Zhang CH, Zhang ZH. Exosomal miR-132-3p from mesenchymal stem cells alleviated LPS-induced acute lung injury by repressing TRAF6. Autoimmunity 2021; 54:493-503. [PMID: 34533429 DOI: 10.1080/08916934.2021.1966768] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Exosomes isolated from mesenchymal stem cells (MSC) had shown beneficial effect on acute lung injury (ALI). However, the effective components in MSC-derived exosomes need further investigation. ALI mice model was established by lipopolysaccharide (LPS) injection. In vitro inflammatory model was established by LPS stimulation of MLE-12 cells. The cell proliferation was evaluated by EdU assay. TUNEL and Annexin V/PI were applied to evaluate the apoptosis of tissue and cell respectively. HE staining was performed to evaluate the lung injury. Transmission electronic microscope was used to observe isolated exosomes. Level of cytokines, MDA, KGF were determined by ELISA kit. Direct interaction of miR-132-3p and TRAF6 were verified by dual luciferase assay. The level of mRNA or proteins were determined by qRT-PCR or western blots respectively. TRAF6 was upregulated while miR-132-3p was downregulated in LPS-stimulated ALI model. MiR-132-3p negatively regulated TRAF6 by direct binding. MiR-132-3p potentiated proliferation and suppressed apoptosis of LPS-induced MLE-12 cells at least partly dependent on targeting TRAF6. Treatment of exosome alleviated the LPS-induced ALI in mice and LPS-induced inflammatory response in MLE-12 cells. Moreover, overexpression of miR-132-3p promoted the protective effect of exosomes in LPS-induced MLE-12 cells injury and LPS-induced ALI. Mechanically, it was suggested that miR-132-3p inactivated PI3K/Akt signalling via targeting TRAF6. In the present study, our results indicated that miR-132-3p mediated protective effect of MSC-derived exosomes on LPS-induced ALI. Exosomal miR-132-3p ameliorated LPS-induced ALI via targeting TRAF6 and inactivating PI3K/Akt signalling.
Collapse
Affiliation(s)
- Jian-Hua Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Hebei North University, Zhangjiakou, P. R. China
| | - Chen Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Hebei North University, Zhangjiakou, P. R. China
| | - Liang Cao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Hebei North University, Zhangjiakou, P. R. China
| | - Chang-Hong Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Hebei North University, Zhangjiakou, P. R. China
| | - Zhi-Hua Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Hebei North University, Zhangjiakou, P. R. China
| |
Collapse
|
42
|
Chen X, Wei Q, Sun H, Zhang X, Yang C, Tao Y, Nong G. Exosomes Derived from Human Umbilical Cord Mesenchymal Stem Cells Regulate Macrophage Polarization to Attenuate Systemic Lupus Erythematosus-Associated Diffuse Alveolar Hemorrhage in Mice. Int J Stem Cells 2021; 14:331-340. [PMID: 33906978 PMCID: PMC8429939 DOI: 10.15283/ijsc20156] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/24/2020] [Accepted: 01/28/2021] [Indexed: 01/01/2023] Open
Abstract
Background and Objectives To investigate the effect and the underlying mechanism of exosomes secreted by human umbilical cord mesenchymal stem cells (hUCMSCs) on diffuse alveolar hemorrhage (DAH) in murine lupus. Methods and Results Exosomes were extracted from cultured hUCMSCs by ultracentrifugation. The expressions of exosome markers (Alix, CD63 and TSG101) were measured for identification of hUCMSC-derived exosomes (hUCMSC-exosomes). The alveolar hemorrhage of DAH mice was revealed by H&E staining. The primary alveolar macrophages were isolated from bronchoalveolar lavage fluid (BALF) of DAH mice. The expressions of M1 macrophage markers (iNOS, IL-6, TNF-α and IL-1β) and M2 macrophage markers (Arg1, IL-10, TGF-β and chi3l3) were detected. Flow cytometry measured the ratio of M1/M2 macrophages. ELISA measured the secretion of pro-inflammatory cytokines (IL-6 and TNF-α) and anti-inflammatory cytokines (IL-10 and TGF-β). DAH mice had hemorrhage and small-vessel vasculitis in the lung, with neutrophil and monocyte infiltration observed around the capillary and small artery. Furthermore, increases of IL-6 and TNF-α, and decreases of IL-10 and TGF-β were detected in the BALF of DAH mice. M1 makers were overexpressed in alveolar macrophages of DAH mice while M2 makers were lowly expressed. DAH mice had a higher proportion of M1 macrophages than M2 macrophages. After hUCMSC-exosome or methylprednisolone treatment in DAH mice, the alveolar injuries and inflammatory responses were attenuated, and the proportion of M2 macrophages was increased. Conclusions hUCMSC-exosomes attenuate DAH-induced inflammatory responses and alveolar hemorrhage by regulating macrophage polarization.
Collapse
Affiliation(s)
- Xun Chen
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Guangxi, China
| | - Qing Wei
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Guangxi, China
| | - Hongmei Sun
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Guangxi, China
| | - Xiaobo Zhang
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Guangxi, China
| | - Changrong Yang
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Guangxi, China
| | - Ying Tao
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Guangxi, China
| | - Guangmin Nong
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Guangxi, China
| |
Collapse
|
43
|
Abdul Halim NSS, Yahaya BH, Lian J. Therapeutic Potential of Adipose-Derived Stem Cells in the Treatment of Pulmonary Diseases. Curr Stem Cell Res Ther 2021; 17:103-112. [PMID: 34387168 DOI: 10.2174/1574888x16666210812145202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 05/07/2021] [Accepted: 05/25/2021] [Indexed: 11/22/2022]
Abstract
Stem cells derived from adipose tissues (ADSCs) have emerged as an ideal candidate for various models of respiratory diseases, including asthma, chronic obstructive pulmonary disease (COPD), and acute respiratory distress syndrome. ADSCs have qualities that may make them better suited for treating inflammatory lung diseases than other MSCs. ADSCs show a lower senescence ratio, higher proliferative capacity and stability in terms of their genetic and morphology during long-term culture over bone marrow-derived mesenchymal stem cells (BMMSCs). With advanced research techniques, the advantageous effects of ADSCs seem limited to their ability to engraft, differentiate, and be related to their secretion of trophic factors. These trophic factors regulate the therapeutic and regenerative outcomes in various lung inflammatory diseases. Taken together, these particular qualities of ADSCs make them significantly relevant for clinical applications. This article discusses a recent advance of ADSCs biology and their translational application emphasizing their anti-inflammatory, immunomodulatory and regenerative properties particularly on lung inflammatory diseases. Besides, the relevant advancements made in the field, the regulatory aspects, and other challenges and obstacles will be highlighted.
Collapse
Affiliation(s)
- Nur Shuhaidatul Sarmiza Abdul Halim
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), SAINS@BERTAM, Universiti Sains Malaysia, 13200 Kepala Batas, Penang . Malaysia
| | - Badrul Hisham Yahaya
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), SAINS@BERTAM, Universiti Sains Malaysia, 13200 Kepala Batas, Penang . Malaysia
| | - Jie Lian
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), SAINS@BERTAM, Universiti Sains Malaysia, 13200 Kepala Batas, Penang . Malaysia
| |
Collapse
|
44
|
Moghadasi S, Elveny M, Rahman HS, Suksatan W, Jalil AT, Abdelbasset WK, Yumashev AV, Shariatzadeh S, Motavalli R, Behzad F, Marofi F, Hassanzadeh A, Pathak Y, Jarahian M. A paradigm shift in cell-free approach: the emerging role of MSCs-derived exosomes in regenerative medicine. J Transl Med 2021; 19:302. [PMID: 34253242 PMCID: PMC8273572 DOI: 10.1186/s12967-021-02980-6] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 07/06/2021] [Indexed: 12/17/2022] Open
Abstract
Recently, mesenchymal stem/stromal cells (MSCs) due to their pro-angiogenic, anti-apoptotic, and immunoregulatory competencies along with fewer ethical issues are presented as a rational strategy for regenerative medicine. Current reports have signified that the pleiotropic effects of MSCs are not related to their differentiation potentials, but rather are exerted through the release of soluble paracrine molecules. Being nano-sized, non-toxic, biocompatible, barely immunogenic, and owning targeting capability and organotropism, exosomes are considered nanocarriers for their possible use in diagnosis and therapy. Exosomes convey functional molecules such as long non-coding RNAs (lncRNAs) and micro-RNAs (miRNAs), proteins (e.g., chemokine and cytokine), and lipids from MSCs to the target cells. They participate in intercellular interaction procedures and enable the repair of damaged or diseased tissues and organs. Findings have evidenced that exosomes alone are liable for the beneficial influences of MSCs in a myriad of experimental models, suggesting that MSC- exosomes can be utilized to establish a novel cell-free therapeutic strategy for the treatment of varied human disorders, encompassing myocardial infarction (MI), CNS-related disorders, musculoskeletal disorders (e.g. arthritis), kidney diseases, liver diseases, lung diseases, as well as cutaneous wounds. Importantly, compared with MSCs, MSC- exosomes serve more steady entities and reduced safety risks concerning the injection of live cells, such as microvasculature occlusion risk. In the current review, we will discuss the therapeutic potential of MSC- exosomes as an innovative approach in the context of regenerative medicine and highlight the recent knowledge on MSC- exosomes in translational medicine, focusing on in vivo researches.
Collapse
Affiliation(s)
- Soudeh Moghadasi
- Department of Biochemistry and Molecular Biology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Marischa Elveny
- DS & CI Research Group, Universitas Sumatera Utara, Medan, Indonesia
| | - Heshu Sulaiman Rahman
- College of Medicine, University of Sulaimani, Sulaymaniyah, Iraq.,Department of Medical Laboratory Sciences, Komar University of Science and Technology, Sulaymaniyah, Iraq
| | - Wanich Suksatan
- Faculty of Nursing, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, 10210, Thailand
| | | | - Walid Kamal Abdelbasset
- Department of Health and Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Al Kharj, Saudi Arabia.,Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza, Egypt
| | | | - Siavash Shariatzadeh
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Roza Motavalli
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farahnaz Behzad
- Research Institute of Bioscience and Biotechnology, University of Tabriz, Tabriz, Iran
| | - Faroogh Marofi
- Department of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Hassanzadeh
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Yashwant Pathak
- Taneja College of Pharmacy, University of South Florida, Tampa Florida, USA
| | - Mostafa Jarahian
- German Cancer Research Center, Toxicology and Chemotherapy Unit (G401), 69120, Heidelberg, Germany.
| |
Collapse
|
45
|
BMSC-Derived Exosomes Ameliorate LPS-Induced Acute Lung Injury by miR-384-5p-Controlled Alveolar Macrophage Autophagy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9973457. [PMID: 34234888 PMCID: PMC8216833 DOI: 10.1155/2021/9973457] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 05/01/2021] [Accepted: 05/26/2021] [Indexed: 12/29/2022]
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are common critical diseases. Bone marrow mesenchymal stem cell (BMSC) transplantation is previously shown to effectively rescue injured lung tissues. The therapeutic mechanism of BMSC-derived exosomes is not fully understood. Here, we investigated the BMSC-derived exosomal microRNAs (miRNAs) on effecting lipopolysaccharide- (LPS-) induced ALI and its mechanism. In vitro, rat alveolar macrophages were treated with or without exosomes in the presence of 10 μg/ml LPS for 24 h. Cell viability was determined with Cell Counting Kit-8 assay. Apoptotic ratio was determined with TUNEL and Annexin V-FITC/PI double staining. The levels of miR-384-5p and autophagy-associated genes were measured by RT-qPCR and western blot. Autophagy was observed by TEM and assessed by means of the mRFP-GFP-LC3 adenovirus transfection assay. In vivo, we constructed LPS-induced ALI rat models. Exosomes were injected into rats via the caudal vein or trachea 4 h later after LPS treatment. The lung histological pathology was determined by H&E staining. Pulmonary vascular permeability was assessed by wet-to-dry weight ratio and Evans blue dye leakage assay, and inflammatory cytokines in serum and BALF were measured by ELISA. Furthermore, the therapeutic mechanism involved in miR-384-5p and Beclin-1 was determined. The results showed that BMSC-derived exosomes were taken up by the alveolar macrophages and attenuated LPS-induced alveolar macrophage viability loss and apoptosis. Exosomes effectively improved the survival rate of ALI rats within 7 days, which was associated with alleviating lung pathological changes and pulmonary vascular permeability and attenuating inflammatory response. Furthermore, this study for the first time found that miR-384-5p was enriched in BMSC-derived exosomes, and exosomal miR-384-5p resulted in relieving LPS-injured autophagy disorder in alveolar macrophages by targeting Beclin-1. Therefore, exosomal miR-384-5p could be demonstrated as a promising therapeutic strategy for ALI/ARDS.
Collapse
|
46
|
Gupta A, Shivaji K, Kadam S, Gupta M, Rodriguez HC, Potty AG, El-Amin SF, Maffulli N. Immunomodulatory extracellular vesicles: an alternative to cell therapy for COVID-19. Expert Opin Biol Ther 2021; 21:1551-1560. [PMID: 33886388 DOI: 10.1080/14712598.2021.1921141] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction: SARS-CoV-2 induces a cytokine storm and can cause inflammation, fibrosis and apoptosis in the lungs, leading to acute respiratory distress syndrome (ARDS). ARDS is the leading cause of mortality and morbidity the associated to COVID-19, and the cytokine storm is a prominent etiological factor. Mesenchymal stem cell-derived extracellular vesicles are an alternative therapy for the management of inflammatory and autoimmune conditions due to their immunosuppressive properties. The immunomodulatory and tissue regeneration capabilities of extracellular vesicles may support their application as a prospective therapy for COVID-19.Areas Covered: We explored the clinical evidence on extracellular vesicles as antiviral agents and in mitigating ARDS, and their therapeutic potential in COVID-19.Expert Opinion: Clinical trials using extracellular vesicles are registered against COVID-19 associated complications, with some evidence of safety and efficacy. Extracellular vesicles present an alternative potential for cell therapy for COVID-19 management, but further preclinical and clinical investigations are needed.
Collapse
Affiliation(s)
- Ashim Gupta
- Future Biologics, Lawrenceville, USA.,BioIntegrate, Lawrenceville, USA.,South Texas Orthopedic Research Institute (STORI Inc), Laredo, USA.,Veterans in Pain, Los Angeles, USA
| | - Kashte Shivaji
- Department of Stem Cell & Regenerative Medicine, Centre for Interdisciplinary Research, D. Y. Patil Education Society (Institution Deemed to Be University), Kolhapur, India
| | - Sachin Kadam
- Department of Stem Cell & Regenerative Medicine, Centre for Interdisciplinary Research, D. Y. Patil Education Society (Institution Deemed to Be University), Kolhapur, India.,Advancells Group, Noida, India
| | | | - Hugo C Rodriguez
- Future Biologics, Lawrenceville, USA.,South Texas Orthopedic Research Institute (STORI Inc), Laredo, USA.,Future Physicians of South Texas, San Antonio, USA.,School of Osteopathic Medicine, University of the Incarnate Word, San Antonio, USA
| | - Anish G Potty
- South Texas Orthopedic Research Institute (STORI Inc), Laredo, USA.,School of Osteopathic Medicine, University of the Incarnate Word, San Antonio, USA.,The Institute of Musculoskeletal Excellence (TIME Orthopaedics), Laredo, USA
| | - Saadiq F El-Amin
- BioIntegrate, Lawrenceville, USA.,El-Amin Orthopaedic & Sports Medicine Institute, Lawrenceville, USA
| | - Nicola Maffulli
- Department of Musculoskeletal Disorders, School of Medicine and Surgery, University of Salerno, Fisciano, Italy.,San Giovanni Di Dio E Ruggi D'Aragona Hospital "Clinica Orthopedica" Department, Hospital of Salerno, Salerno, Italy.,Queen Mary University of London, Barts and the London School of Medicine and Dentistry, Centre for Sports and Exercise Medicine, London, England
| |
Collapse
|
47
|
d’Alessandro M, Soccio P, Bergantini L, Cameli P, Scioscia G, Foschino Barbaro MP, Lacedonia D, Bargagli E. Extracellular Vesicle Surface Signatures in IPF Patients: A Multiplex Bead-Based Flow Cytometry Approach. Cells 2021; 10:cells10051045. [PMID: 33925174 PMCID: PMC8146446 DOI: 10.3390/cells10051045] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/14/2021] [Accepted: 04/26/2021] [Indexed: 12/20/2022] Open
Abstract
Background: Extracellular vesicles (EVs) are secreted by cells from their membrane within circulation and body fluids. Knowledge of the involvement of EVs in pathogenesis of lung diseases is increasing. The present study aimed to evaluate the expression of exosomal surface epitopes in a cohort of idiopathic pulmonary fibrosis (IPF) patients followed in two Italian Referral Centres for Interstitial Lung Diseases, comparing them with a group of healthy volunteers. Materials and Methods: Ninety IPF patients (median age and interquartile range (IQR) 71 (66–75) years; 69 males) were selected retrospectively. Blood samples were obtained from patients before starting antifibrotic therapy. A MACSPlex Exosome Kit, human, (Miltenyi Biotec, Bergisch-Gladbach, Germany), to detect 37 exosomal surface epitopes, was used. Results: CD19, CD69, CD8, and CD86 were significantly higher in IPF patients than in controls (p = 0.0023, p = 0.0471, p = 0.0082, and p = 0.0143, respectively). CD42a was lower in IPF subjects than in controls (p = 0.0153), while CD209, Cd133/1, MCSP, and ROR1 were higher in IPF patients than in controls (p = 0.0007, p = 0.0050, p = 0.0139, and p = 0.0335, respectively). Kaplan-Meier survival analysis for IPF patients: for median values and a cut-off of 0.48 for CD25, the two subgroups showed a significant difference in survival rate (p = 0.0243, hazard ratio: 0.52 (95%CI 0.29–0.92); the same was true for CD8 (cut-off 1.53, p = 0.0309, hazard ratio: 1.39 (95%CI 0.75–2.53). Conclusion: Our multicenter study showed for the first time the expression of surface epitopes on EVs from IPF patients, providing interesting data on the communication signatures/exosomal profile in serum from IPF patients and new insights into the pathogenesis of the disease and a promising reliability in predicting mid-term survival of IPF patients.
Collapse
Affiliation(s)
- Miriana d’Alessandro
- Respiratory Diseases Unit, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, 53100 Siena, Italy; (L.B.); (P.C.); (E.B.)
- Correspondence: ; Tel.: +39-057-758-6713; Fax: +39-057-728-0744
| | - Piera Soccio
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (P.S.); (G.S.); (M.P.F.B.); (D.L.)
- Institute of Respiratory Diseases, Policlinico Riuniti of Foggia, 71122 Foggia, Italy
| | - Laura Bergantini
- Respiratory Diseases Unit, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, 53100 Siena, Italy; (L.B.); (P.C.); (E.B.)
| | - Paolo Cameli
- Respiratory Diseases Unit, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, 53100 Siena, Italy; (L.B.); (P.C.); (E.B.)
| | - Giulia Scioscia
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (P.S.); (G.S.); (M.P.F.B.); (D.L.)
- Institute of Respiratory Diseases, Policlinico Riuniti of Foggia, 71122 Foggia, Italy
| | - Maria Pia Foschino Barbaro
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (P.S.); (G.S.); (M.P.F.B.); (D.L.)
- Institute of Respiratory Diseases, Policlinico Riuniti of Foggia, 71122 Foggia, Italy
| | - Donato Lacedonia
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (P.S.); (G.S.); (M.P.F.B.); (D.L.)
- Institute of Respiratory Diseases, Policlinico Riuniti of Foggia, 71122 Foggia, Italy
| | - Elena Bargagli
- Respiratory Diseases Unit, Department of Medical and Surgical Sciences & Neurosciences, Siena University Hospital, 53100 Siena, Italy; (L.B.); (P.C.); (E.B.)
| |
Collapse
|
48
|
Jamalkhah M, Asaadi Y, Azangou-Khyavy M, Khanali J, Soleimani M, Kiani J, Arefian E. MSC-derived exosomes carrying a cocktail of exogenous interfering RNAs an unprecedented therapy in era of COVID-19 outbreak. J Transl Med 2021; 19:164. [PMID: 33888147 PMCID: PMC8061879 DOI: 10.1186/s12967-021-02840-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 04/16/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The onset of the SARS-CoV-2 pandemic has resulted in ever-increasing casualties worldwide, and after 15 months, standard therapeutic regimens are yet to be discovered. MAIN BODY Due to the regenerative and immunomodulatory function of MSCs, they can serve as a suitable therapeutic option in alleviating major COVID-19 complications like acute respiratory distress syndrome. However, the superior properties of their cognate exosomes as a cell-free product make them preferable in the clinic. Herein, we discuss the current clinical status of these novel therapeutic strategies in COVID-19 treatment. We then delve into the potential of interfering RNAs incorporation as COVID-19 gene therapy and introduce targets involved in SARS-CoV-2 pathogenesis. Further, we present miRNAs and siRNAs candidates with promising results in targeting the mentioned targets. CONCLUSION Finally, we present a therapeutic platform of mesenchymal stem cell-derived exosomes equipped with exogenous iRNAs, that can be employed as a novel therapeutic modality in COVID-19 management aiming to prevent further viral spread within the lung, hinder the virus life cycle and pathogenesis such as immune suppression, and ultimately, enhance the antiviral immune response.
Collapse
Affiliation(s)
- Monire Jamalkhah
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Yasaman Asaadi
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | | | - Javad Khanali
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoud Soleimani
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jafar Kiani
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Ehsan Arefian
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran.
| |
Collapse
|
49
|
Li KL, Li JY, Xie GL, Ma XY. Exosomes Released From Human Bone Marrow-Derived Mesenchymal Stem Cell Attenuate Acute Graft-Versus-Host Disease After Allogeneic Hematopoietic Stem Cell Transplantation in Mice. Front Cell Dev Biol 2021; 9:617589. [PMID: 33889570 PMCID: PMC8055957 DOI: 10.3389/fcell.2021.617589] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 02/08/2021] [Indexed: 12/14/2022] Open
Abstract
Objective Mesenchymal stromal cell–derived exosomes have been applied for the treatment of several immune diseases. This study aimed to explore the effect of human bone marrow–derived mesenchymal stem cell (hBMSC)–derived exosomes on acute graft-versus-host disease (aGVHD) after allogeneic hematopoietic stem cell transplantation (HSCT). Methods hBMSC were cultured, and the culture supernatants were then collected to prepare exosomes using total exosome isolation reagent from Invitrogen. Mouse aGVHD model was established by allogeneic cell transplantation and injected with hBMSC-derived exosomes (Msc-exo) via tail vein. Exosomes from human fibroblast (Fib-exo) were used as the treatment control. The effects of Msc-exo on dendritic cells, CD4+, and CD8+ T cells in aGVHD mice were analyzed through flow cytometry. The impact on inflammatory cytokines was tested by ELISA. Besides, the body weight, survival rate, and clinical score of treated mice were monitored. Results Msc-exo were successfully prepared. aGVHD mice injected with Msc-exo led to 7–8-fold increase of the CD8α+ conventional dendritic cells (cDCs) and CD11b+ cDCs compared with the controls. In addition, Msc-exo altered the T help and Treg subpopulation, and decreased the cytotoxicity and proliferation of cytotoxic T cells to favor inflammatory inhibition in aGVHD mice. Mice that received Msc-exo exhibited decreased weight loss and reduced aGVHD clinical score in a time-dependent manner as well as reduced lethality compared with Fib-exo treated or untreated control. Furthermore, the levels of IL-2, TNF-α, and IFN-γ were decreased, as well as the level of IL-10 was increased after Msc-exo treatment in vivo and in vitro. Conclusion hBMSC-derived exosomes could attenuate aGVHD damage and promote the survival of aGVHD mice by regulating the DC and T-cell subpopulation and function, and lead to inhibited inflammatory response in aGVHD mice.
Collapse
Affiliation(s)
- Ke-Liang Li
- Department of Pediatrics, Rizhao People's Hospital, Rizhao, China
| | - Jin-Yan Li
- Department of Pediatrics, Rizhao People's Hospital, Rizhao, China
| | - Gui-Ling Xie
- Department of Pediatrics, Rizhao People's Hospital, Rizhao, China
| | - Xiao-Yan Ma
- Department of Pediatrics, Rizhao People's Hospital, Rizhao, China
| |
Collapse
|
50
|
Peng W, Chang M, Wu Y, Zhu W, Tong L, Zhang G, Wang Q, Liu J, Zhu X, Cheng T, Li Y, Chen X, Weng D, Liu S, Zhang H, Su Y, Zhou J, Li H, Song Y. Lyophilized powder of mesenchymal stem cell supernatant attenuates acute lung injury through the IL-6-p-STAT3-p63-JAG2 pathway. Stem Cell Res Ther 2021; 12:216. [PMID: 33781349 PMCID: PMC8008635 DOI: 10.1186/s13287-021-02276-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 03/10/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are syndromes of acute respiratory failure with extremely high mortality and few effective treatments. Mesenchymal stem cells (MSCs) may reportedly contribute to tissue repair in ALI and ARDS. However, applications of MSCs have been restricted due to safety considerations and limitations in terms of large-scale production and industrial delivery. Alternatively, the MSC secretome has been considered promising for use in therapeutic approaches and has been advanced in pre-clinical and clinical trials. Furthermore, the MSC secretome can be freeze-dried into a stable and ready-to-use supernatant lyophilized powder (SLP) form. Currently, there are no studies on the role of MSC SLP in ALI. METHODS Intratracheal bleomycin was used to induce ALI in mice, and intratracheal MSC SLP was administered as a treatment. Histopathological assessment was performed by hematoxylin and eosin, immunohistochemistry, and immunofluorescence staining. Apoptosis, inflammatory infiltration, immunological cell counts, cytokine levels, and mRNA- and protein-expression levels of relevant targets were measured by performing terminal deoxynucleotidyl transferase dUTP nick-end labeling assays, determining total cell and protein levels in bronchoalveolar lavage fluids, flow cytometry, multiple cytokine-detection techniques, and reverse transcriptase-quantitative polymerase chain reaction and western blot analysis, respectively. RESULTS We found that intratracheal MSC SLP considerably promoted cell survival, inhibited epithelial cell apoptosis, attenuated inflammatory cell recruitment, and reversed immunological imbalances induced by bleomycin. MSC SLP inhibited the interleukin 6-phosphorylated signal transducer and activator of transcription signaling pathway to activate tumor protein 63-jagged 2 signaling in basal cells, suppress T helper 17 cell differentiation, promote p63+ cell proliferation and lung damage repair, and attenuate inflammatory responses. CONCLUSIONS MSC SLP ameliorated ALI by activating p63 and promoting p63+ cell proliferation and the repair of damaged epithelial cells. The findings of this study also shed insight into ALI pathogenesis and imply that MSC SLP shows considerable therapeutic promise for treating ALI and ARDS.
Collapse
Affiliation(s)
- Wenjun Peng
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Meijia Chang
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yuanyuan Wu
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wensi Zhu
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Lin Tong
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ge Zhang
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Qin Wang
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jie Liu
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiaoping Zhu
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Tingting Cheng
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yijia Li
- Public Translational Platform for Cell Therapy, Yangtze Delta Region Institute of Tsinghua University, Hangzhou, 311200, Zhejiang, China
| | - Xi Chen
- Yunnan Province Stem cell Bank, Kunming, 650101, Yunnan, China
| | - Dong Weng
- Department of Pulmonary and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Sanhong Liu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Hongwei Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yao Su
- Public Translational Platform for Cell Therapy, Yangtze Delta Region Institute of Tsinghua University, Hangzhou, 311200, Zhejiang, China
| | - Jian Zhou
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China. .,Center of Emergency & Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, 200540, China.
| | - Huayin Li
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Yuanlin Song
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China. .,Center of Emergency & Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, 200540, China.
| |
Collapse
|