1
|
Ding Z, Ma G, Zhou B, Cheng S, Tang W, Han Y, Chen L, Pang W, Chen Y, Yang D, Cao H. Targeting miR-29 mitigates skeletal senescence and bolsters therapeutic potential of mesenchymal stromal cells. Cell Rep Med 2024; 5:101665. [PMID: 39168101 PMCID: PMC11384963 DOI: 10.1016/j.xcrm.2024.101665] [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: 03/05/2024] [Revised: 06/07/2024] [Accepted: 07/08/2024] [Indexed: 08/23/2024]
Abstract
Mesenchymal stromal cell (MSC) senescence is a key factor in skeletal aging, affecting the potential of MSC applications. Identifying targets to prevent MSC and skeletal senescence is crucial. Here, we report increased miR-29 expression in bone tissues of aged mice, osteoporotic patients, and senescent MSCs. Genetic overexpression of miR-29 in Prx1-positive MSCs significantly accelerates skeletal senescence, reducing cortical bone thickness and trabecular bone mass, while increasing femur cross-sectional area, bone marrow adiposity, p53, and senescence-associated secretory phenotype (SASP) levels. Mechanistically, miR-29 promotes senescence by upregulating p53 via targeting Kindlin-2 mRNA. miR-29 knockdown in BMSCs impedes skeletal senescence, enhances bone mass, and accelerates calvarial defect regeneration, also reducing lipopolysaccharide (LPS)-induced organ injuries and mortality. Thus, our findings underscore miR-29 as a promising therapeutic target for senescence-related skeletal diseases and acute inflammation-induced organ damage.
Collapse
Affiliation(s)
- Zhen Ding
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Guixing Ma
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China.
| | - Bo Zhou
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Siyuan Cheng
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Wanze Tang
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yingying Han
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Litong Chen
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Wei Pang
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yangshan Chen
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Dazhi Yang
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Huiling Cao
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China.
| |
Collapse
|
2
|
Ma N, Zhang M, Xu G, Zhang L, Luo M, Luo M, Wang X, Tang H, Wang X, Liu L, Zhong X, Feng J, Li Y. Mesenchymal Stem Cell-derived Type II Alveolar Epithelial Progenitor Cells Attenuate LPS-induced Acute Lung Injury and Reduce P63 Expression. Curr Stem Cell Res Ther 2024; 19:245-256. [PMID: 37138488 DOI: 10.2174/1574888x18666230501234836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/26/2023] [Accepted: 02/28/2023] [Indexed: 05/05/2023]
Abstract
AIM Acute respiratory distress syndrome (ARDS)/acute lung injury (ALI) is a severe clinical respiratory-failure disease mainly characterized by acute damage to the alveolar epithelium and pulmonary vascular endothelial cells. Stem cell therapy has emerged as a potential regenerative strategy for ARDS/ALI, however, the outcome is limited, and the underlying mechanisms are unclear. INTRODUCTION We established a differentiation system for bone marrow-derived mesenchymal stem cellderived (BM-MSC) type II alveolar epithelial progenitor cells (AECIIs) and assessed their regulatory effects on lipopolysaccharide (LPS)-induced ALI. METHODS We induced BM-MSC differentiation into AECIIs using a specific conditioned medium. After 26 days of differentiation, 3×105 BM-MSC-AECIIs were used to treat mice with LPS-induced ALI through tracheal injection. RESULTS After tracheal injection, BM-MSC-AECIIs migrated to the perialveolar area and reduced LPSinduced lung inflammation and pathological injury. RNA-seq suggested that P63 protein was involved in the effects of BM-MSC-AECIIs on lung inflammation. CONCLUSION Our results suggest that BM-MSC-AECIIs may reduce LPS-induced acute lung injury by decreasing P63 expression.
Collapse
Affiliation(s)
- Ning Ma
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Mengwei Zhang
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Guofeng Xu
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Lifang Zhang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Min Luo
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Meihua Luo
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xing Wang
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Hongmei Tang
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xiaoyun Wang
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Li Liu
- Laboratory of Anesthesiology, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xiaolin Zhong
- Department of Gastroenterology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Jianguo Feng
- Laboratory of Anesthesiology, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Yuying Li
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| |
Collapse
|
3
|
Zhang S, Hu D, Zhuo Y, Cui L, Li D, Zhang L, Yang L, Wang X. Protective effect of liriodendrin on IgG immune complex-induced acute lung injury via inhibiting SRC/STAT3/MAPK signaling pathway: a network pharmacology research. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:3269-3283. [PMID: 37243760 DOI: 10.1007/s00210-023-02534-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 05/16/2023] [Indexed: 05/29/2023]
Abstract
The primary objectives of this research were to investigate the protective effects of liriodendrin against IgG immune complex (IgG-IC)-induced acute lung injury (ALI) and to elucidate the underlying mechanisms. This study employed a mouse and cell model of IgG-IC-induced acute lung injury. Lung tissue was stained with hematoxylin-eosin to observe pathological alterations and arterial blood gas analysis was tested. Inflammatory cytokines, including interleukin-6 (IL-6), interleukin-1β (IL-1β), and tumor necrosis factor-alpha (TNF-α), were measured using ELISA. The mRNA expression of inflammatory cytokines was assessed via RT-qPCR. Molecular docking and enrichment analysis were combined to identify the most potential signaling pathways modulated by liriodendrin, which were then verified using western blot analysis in IgG-IC-induced ALI models. We identified 253 shared targets between liriodendrin and IgG-IC-induced acute lung injury from the database. Through network pharmacology, enrichment analysis, and molecular docking, SRC was determined to be the most closely associated target of liriodendrin in IgG-IC-induced ALI. Pretreatment with liriodendrin notably reduced the increased cytokine secretion of IL-1β, IL-6, and TNF-α. Histopathological analysis of lung tissue demonstrated a protective effect of liriodendrin on IgG-IC-induced acute lung injury in mice. Arterial blood gas analysis showed liriodendrin ameliorated acidosis and hypoxemia efficiently. Further studies revealed that liriodendrin pretreatment substantially attenuated the elevated phosphorylation levels of SRC's downstream components (JNK, P38, and STAT3), suggesting that liriodendrin may protect against IgG-IC-induced ALI via the SRC/STAT3/MAPK pathway. Our findings indicate that liriodendrin protects against IgG-IC-induced acute lung injury by inhibiting the SRC/STAT3/MAPK signaling pathway, suggesting that liriodendrin may serve as a potential treatment for acute lung injury caused by IgG-IC.
Collapse
Affiliation(s)
- Sijia Zhang
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Dongsheng Hu
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Yuzhen Zhuo
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, China
| | - Lingzhi Cui
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Dihua Li
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, China
| | - Lanqiu Zhang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, China
| | - Lei Yang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, China.
| | - Ximo Wang
- Graduate School, Tianjin Medical University, Tianjin, China.
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Nankai Hospital, Tianjin, China.
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Integrative Medicine for Acute Abdominal Diseases, Tianjin University, Tianjin, China.
| |
Collapse
|
4
|
Zhao J, Liang Q, Fu C, Cong D, Wang L, Xu X. Autophagy in sepsis-induced acute lung injury: Friend or foe? Cell Signal 2023; 111:110867. [PMID: 37633477 DOI: 10.1016/j.cellsig.2023.110867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/14/2023] [Accepted: 08/23/2023] [Indexed: 08/28/2023]
Abstract
Sepsis-induced acute lung injury (ALI) is a life-threatening syndrome with high mortality and morbidity, resulting in a heavy burden on family and society. As a key factor that maintains cellular homeostasis, autophagy is regarded as a self-digesting process by which damaged organelles and useless proteins are recycled for cell metabolism, and it thus plays a crucial role during physiological and pathological processes. Recent studies have indicated that autophagy is involved in the pathophysiological process of sepsis-induced ALI, including cell apoptosis, inflammation, and mitochondrial dysfunction, which indicates that regulating autophagy may be beneficial for this disease. However, the role of autophagy in the etiology and treatment of sepsis-induced ALI is not well characterized. This review summarizes the autophagy-related signaling pathways in sepsis-induced ALI, as well as focuses on the dual role of autophagy and its regulation by non-coding RNAs during disease progression, for the development of potential therapeutic strategies in this disease.
Collapse
Affiliation(s)
- Jiayao Zhao
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Qun Liang
- Department of Critical Care Medicine, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Chenfei Fu
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Didi Cong
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Long Wang
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Xiaoxin Xu
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin 150040, China.
| |
Collapse
|
5
|
Zhang S, Yang L, Hu D, He S, Cui L, Zhao J, Zhuo Y, Zhang L, Wang X. Syringaresinol alleviates IgG immune complex induced acute lung injury via activating PPARγ and suppressing pyroptosis. Int Immunopharmacol 2023; 124:111071. [PMID: 37857123 DOI: 10.1016/j.intimp.2023.111071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 10/05/2023] [Accepted: 10/10/2023] [Indexed: 10/21/2023]
Abstract
Acute lung injury (ALI) is a life-threatening condition characterized by severe lung inflammation and tissue damage. In this study, we investigate the potential therapeutic efficacy of (+)-Syringaresinol (SYG), a natural compound known for its antioxidant and anti-inflammatory properties, in alleviating ALI induced by IgG immune complexes (IgG-IC). Using MH-S cells as a model, we explore SYG's ability to target peroxisome proliferator-activated receptor gamma (PPARγ) and its anti-inflammatory properties. Our comprehensive investigation aims to elucidate the specific molecular mechanisms underlying SYG's effects against pyroptosis, as revealed through transcriptomic analysis. Validation in C57BL/6 mice provides in vivo support. Our findings indicate that SYG effectively mitigates IgG-IC-induced lung damage, as evidenced by a significant reduction in lung inflammation and tissue injury. SYG treatment notably decreases pro-inflammatory cytokine levels (TNF-α, IL-6, IL-1β) in both lung tissue and cells. Molecular docking analysis reveals SYG's robust binding to PPARγ, leading to the inhibition of IgG-IC-induced inflammatory signaling pathways. Additionally, transcriptomic analysis unveils SYG's potential in suppressing macrophage pyroptosis, potentially through the downregulation of key inflammatory mediators (NLRP3, GSDMD, Caspase-1). In summary, our study presents compelling evidence supporting SYG as an effective therapeutic agent for ALI. SYG's activation of PPARγ contributes to the suppression of NF-κB and C/EBPs expression, thereby mitigating inflammation. Moreover, SYG demonstrates the ability to inhibit macrophage pyroptosis by targeting the NLRP3/GSDMD/caspase-1 axis.
Collapse
Affiliation(s)
- Sijia Zhang
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Lei Yang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Integrated Traditional Chinese and Western Medicine Hospital, Tianjin Nankai Hospital, Tianjin University, Tianjin, China
| | - Dongsheng Hu
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Siqi He
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Lingzhi Cui
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Jiuling Zhao
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Integrated Traditional Chinese and Western Medicine Hospital, Tianjin Nankai Hospital, Tianjin University, Tianjin, China
| | - Yuzhen Zhuo
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Integrated Traditional Chinese and Western Medicine Hospital, Tianjin Nankai Hospital, Tianjin University, Tianjin, China.
| | - Lanqiu Zhang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Integrated Traditional Chinese and Western Medicine Hospital, Tianjin Nankai Hospital, Tianjin University, Tianjin, China.
| | - Ximo Wang
- Graduate School, Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Integrated Traditional Chinese and Western Medicine Hospital, Tianjin Nankai Hospital, Tianjin University, Tianjin, China.
| |
Collapse
|
6
|
Cai B, Song W, Chen S, Sun J, Zhou R, Han Z, Wan J. Bone Mesenchymal Stem Cell-Derived Small Extracellular Vesicles Ameliorated Lipopolysaccharide-Induced Lung Injury Via the miR-21-5p/PCSK6 Pathway. J Immunol Res 2023; 2023:3291137. [PMID: 37937296 PMCID: PMC10626970 DOI: 10.1155/2023/3291137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/18/2023] [Accepted: 09/28/2023] [Indexed: 11/09/2023] Open
Abstract
Acute lung injury (ALI) is a life-threatening disease that currently lacks a cure. Although stem cell-derived small extracellular vesicles (sEVs) have shown promising effects in the treatment of ALI, their underlying mechanisms and responsible components have yet to be identified. Proprotein convertase subtilisin/kexin type 6 (PCSK6) is a gene involved in inflammation and a potential target of miR-21-5p, a microRNA enriched in stem cell-derived sEVs. The current study investigated the role of PCSK6 in lipopolysaccharide (LPS)-induced ALI and its interaction with miR-21-5p. Notably, our results showed that PCSK6 expression was positively correlated with LPS stimulation. Knockdown of PCSK6 ameliorated LPS-induced inhibition of proliferation and upregulation of permeability in human BEAS-2B cells, whereas PCSK6 overexpression displayed the opposite effects. BEAS-2B cells were able to actively internalize the cocultured bone mesenchymal stem cell (MSC)-derived sEVs (BMSC-sEVs), which alleviated the cell damage caused by LPS. Overexpressing PCSK6, however, eliminated the therapeutic effects of BMSC-sEV coculture. Mechanistically, BMSC-sEVs inhibited PCSK6 expression via the delivery of miR-21-5p, which is directly bound to the PCSK6 gene. Our work provides evidence for the role of PCSK6 in LPS-induced ALI and identified miR-21-5p as a component of BMSC-derived sEVs that suppressed PCSK6 expression and ameliorated LPS-induced cell damage. These results reveal a novel molecular mechanism for ALI pathogenesis and highlight the therapeutic potential of using sEVs released by stem cells to deliver miR-21-5p for ALI treatment.
Collapse
Affiliation(s)
- Bo Cai
- Department of Emergency and Critical Care Medicine, Shanghai Pudong New Area People's Hospital, No. 490 Chuansha South Road, Pudong New Area, 201299, Shanghai, China
| | - Weidong Song
- Department of Emergency and Critical Care Medicine, Shanghai Pudong New Area People's Hospital, No. 490 Chuansha South Road, Pudong New Area, 201299, Shanghai, China
| | - Song Chen
- Department of Emergency and Critical Care Medicine, Shanghai Pudong New Area People's Hospital, No. 490 Chuansha South Road, Pudong New Area, 201299, Shanghai, China
| | - Jie Sun
- Department of Emergency and Critical Care Medicine, Shanghai Pudong New Area People's Hospital, No. 490 Chuansha South Road, Pudong New Area, 201299, Shanghai, China
| | - Rui Zhou
- Department of Emergency and Critical Care Medicine, Shanghai Pudong New Area People's Hospital, No. 490 Chuansha South Road, Pudong New Area, 201299, Shanghai, China
| | - Zhen Han
- Department of Emergency and Critical Care Medicine, Shanghai Pudong New Area People's Hospital, No. 490 Chuansha South Road, Pudong New Area, 201299, Shanghai, China
| | - Jian Wan
- Department of Emergency and Critical Care Medicine, Shanghai Pudong New Area People's Hospital, No. 490 Chuansha South Road, Pudong New Area, 201299, Shanghai, China
| |
Collapse
|
7
|
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: 6] [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
|
8
|
Kalhori MR, Soleimani M, Alibakhshi R, Kalhori AA, Mohamadi P, Azreh R, Farzaei MH. The Potential of miR-21 in Stem Cell Differentiation and its Application in Tissue Engineering and Regenerative Medicine. Stem Cell Rev Rep 2023; 19:1232-1251. [PMID: 36899116 DOI: 10.1007/s12015-023-10510-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2023] [Indexed: 03/12/2023]
Abstract
MicroRNAs (miRNAs) and long non-coding RNAs (lncRNAs) are two important types of non-coding RNAs that are not translated into protein. These molecules can regulate various biological processes, including stem cell differentiation and self-renewal. One of the first known miRNAs in mammals is miR-21. Cancer-related studies have shown that this miRNA has proto-oncogene activity and is elevated in cancers. However, it is confirmed that miR-21 inhibits stem cell pluripotency and self-renewal and induces differentiation by targeting various genes. Regenerative medicine is a field of medical science that tries to regenerate and repair damaged tissues. Various studies have shown that miR-21 plays an essential role in regenerative medicine by affecting stem cell proliferation and differentiation. In this review, we will discuss the function of miR-21 in regenerative medicine of the liver, nerve, spinal cord, wound, bone, and dental tissues. In addition, the function of natural compounds and lncRNAs will be analyzed as potential regulators of miR-21 expression in regenerative medicine.
Collapse
Affiliation(s)
- Mohammad Reza Kalhori
- Regenerative Medicine Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Masoud Soleimani
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Reza Alibakhshi
- Department of Biochemistry, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Amir Ali Kalhori
- Regenerative Medicine Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Parisa Mohamadi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical, Sciences, Tehran, Iran
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Rasoul Azreh
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Hosien Farzaei
- Medical Technology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
9
|
Li YL, Qin SY, Li Q, Song SJ, Xiao W, Yao GD. Jinzhen Oral Liquid alleviates lipopolysaccharide-induced acute lung injury through modulating TLR4/MyD88/NF-κB pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 114:154744. [PMID: 36934667 DOI: 10.1016/j.phymed.2023.154744] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 02/12/2023] [Accepted: 03/04/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Acute lung injury (ALI) has the attribution of excessive inflammation of the lung. Jinzhen oral liquid (JO), a famous Chinese recipe used to treat ALI, has a favorable therapeutic effect on ALI. However, its anti-inflammatory mechanism has not been extensively studied. PURPOSE This study was to elucidate the effects of JO on lipopolysaccharide (LPS)-induced ALI and its molecular mechanism. METHODS An ALI model was established by intratracheal instillation of LPS (2 mg/50 μl). The open field experiment was carried out to explore the spontaneous movement and exploratory behavior of ALI mice. Cytokines levels concentrations (IL-6, IL-10 and TNF-α) were determined by enzyme-linked immunosorbent assay (ELISA). Network pharmacology was used to predict the mechanism of JO against ALI. Immunofluorescence, co-immunoprecipitation, fluorescence resonance energy transfer (FRET), Western blot and RT-PCR were used to verify the molecular mechanisms of JO. RESULTS The in vivo results suggested that JO (1, 2, 4 g/kg) dose-dependently improved the exercise performance of mice and reduced the lung W/D weight ratio as well as the production of IL-6 and TNF-α, but increased the release of IL-10 in the ALI group. The network pharmacological analysis demonstrated that the Toll-like receptor (TLR) pathway might be the fundamental action mechanisms of JO against ALI. Immunofluorescence staining and co-immunoprecipitation analysis showed that JO decreased the expression levels of TLR4 and MyD88 and reduced their interaction in the lung tissue of ALI mice. Meanwhile, JO decreased nuclear translocation and phosphorylation of NF-κB P65. The results from cellular experiments were in line with those in vivo. The FRET experiment also confirmed that JO disturbed the interaction of TLR4 and MyD88. Subsequently, we also found that the six indicative components of JO have the similar therapeutic effect as JO. CONCLUSIONS In summary, we suggested that JO suppressed the TLR4/MyD88/NF-κB signaling pathway, thus inhibiting LPS-induced ALI in vitro and in vivo. The clarified mechanism provided an important theoretical basis and a novel treatment strategy for the ALI treatment of JO.
Collapse
Affiliation(s)
- Ya-Ling Li
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province, Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Shu-Yan Qin
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province, Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Qian Li
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province, Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Shao-Jiang Song
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province, Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China.
| | - Wei Xiao
- State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, Jiangsu 222001, China.
| | - Guo-Dong Yao
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, Engineering Research Center of Natural Medicine Active Molecule Research & Development, Liaoning Province, Key Laboratory of Natural Bioactive Compounds Discovery & Modification, Shenyang; School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China; State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, Jiangsu 222001, China.
| |
Collapse
|
10
|
Li R, Ren T, Zeng J, Xu H. ALCAM Deficiency Alleviates LPS-Induced Acute Lung Injury by Inhibiting Inflammatory Response. Inflammation 2023; 46:688-699. [PMID: 36418761 DOI: 10.1007/s10753-022-01765-3] [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] [Received: 03/03/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 11/25/2022]
Abstract
We investigated the effects and underlying mechanisms of activated leukocyte adhesion molecule (ALCAM) on acute lung injury (ALI) by using lipopolysaccharide (LPS)-induced ALI animal model and LPS-induced inflammation in vitro. In LPS-stimulated mice, ALCAM deficiency relieved lung injury, which manifested as reduced pathological changes in the lung tissue, reduced pulmonary edema, and reduced vascular permeability. Furthermore, we demonstrated that ALCAM deficiency reduced the infiltration of inflammatory cells, including neutrophil, eosinophil, and macrophages; the release of inflammatory cytokines, including IL-1β, IL-6, TNF-α, and COX2; and reduced the protein level of TLR4/NF-κB pathway (TLR4, MyD88, p-IkBɑ, and p-NF-κB p65). We also demonstrated that ALCAM deficiency reduced the expression of oxidative stress-related proteins (Nrf-2, HO-1, and NQO-1) and endoplasmic reticulum stress-related proteins (CHOP, GRP78, ATF-6, and p-eIF2ɑ). In addition, in LPS-induced inflammation in vitro, ALCAM overexpression promoted inflammatory response, oxidative stress, and ER stress. We established that ALCAM deficiency can suppress the ALI process by reducing inflammatory response, oxidative stress, and endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Ruirui Li
- Department of Critical Care Medicine, The First Affiliated Hospital of Shihezi University School of Medicine, No. 107, Shibei 2Nd Road, Shihezi, 832008, Xinjiang Uygur Autonomous Region, People's Republic of China
| | - Tao Ren
- Three Departments of Cardiology, The First Affiliated Hospital of Shihezi University School of Medicine, Shihezi, 832008, Xinjiang Uygur Autonomous Region, People's Republic of China
| | - Jianqiong Zeng
- Cardiovascular Surgery CCU, Foshan First People's Hospital, Foshan, 528000, Guangdong, People's Republic of China
| | - Hang Xu
- Department of Critical Care Medicine, The First Affiliated Hospital of Shihezi University School of Medicine, No. 107, Shibei 2Nd Road, Shihezi, 832008, Xinjiang Uygur Autonomous Region, People's Republic of China.
| |
Collapse
|
11
|
Mitophagy-promoting miR-138-5p promoter demethylation inhibits pyroptosis in sepsis-associated acute lung injury. Inflamm Res 2023; 72:329-346. [PMID: 36538076 DOI: 10.1007/s00011-022-01675-y] [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: 07/25/2022] [Revised: 07/25/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The present study was designed to explore the potential regulatory mechanism between mitophagy and pyroptosis during sepsis-associated acute lung injury (ALI). METHODS In vitro or in vivo models of sepsis-associated ALI were established by administering lipopolysaccharide (LPS) or performing caecal ligation and puncture (CLP) surgery. Pyroptosis levels were detected by electron microscopy, immunofluorescence, flow cytometry, western blotting and immunohistochemistry. Dual-luciferase reporter gene assay was applied to verify the targeting relationship between miR-138-5p and NLRP3. Methylation-specific PCR and chromatin immunoprecipitation assays were used to determine methylation of the miR-138-5p promoter. Mitophagy levels were examined by transmission electron microscopy and western blotting. RESULTS NLRP3 inflammasome silencing alleviated alveolar macrophage (AM) pyroptosis and septic lung injury. In addition, we confirmed the direct targeting relationship between miR-138-5p and NLRP3. Overexpressed miR-138-5p alleviated AM pyroptosis and the pulmonary inflammatory response. Moreover, the decreased expression of miR-138-5p was confirmed to depend on promoter methylation, while inhibition of miR-138-5p promoter methylation attenuated AM pyroptosis and pulmonary inflammation. Here, we discovered that an increased cytoplasmic mtDNA content in sepsis-induced ALI models induced the methylation of the miR-138-5p promoter, thereby decreasing miR-138-5p expression, which may activate the NLRP3 inflammasome and trigger AM pyroptosis. Mitophagy, a form of selective autophagy that clears damaged mitochondria, reduced cytoplasmic mtDNA levels. Furthermore, enhanced mitophagy might suppress miR-138-5p promoter methylation and relieve the pulmonary inflammatory response, changes that were reversed by treatment with isolated mtDNA. CONCLUSIONS In summary, our study indicated that mitophagy induced the demethylation of the miR-138-5p promoter, which may subsequently inhibit NLRP3 inflammasome, AM pyroptosis and inflammation in sepsis-induced lung injury. These findings may provide a promising therapeutic target for sepsis-associated ALI.
Collapse
|
12
|
Nrf2 regulates the expression of NOX1 in TNF-α-induced A549 cells. Allergol Immunopathol (Madr) 2023; 51:54-62. [PMID: 36617822 DOI: 10.15586/aei.v51i1.732] [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: 07/15/2022] [Accepted: 09/23/2022] [Indexed: 01/03/2023]
Abstract
Acute lung injury causes severe inflammation and oxidative stress in lung tissues. In this study, we analyzed the potential regulatory role of nuclear factor erythroid-2-related factor 2 (Nrf2) on NADPH oxidase 1 (NOX1) in tumor necrosis factor-α (TNF-α)-induced inflammation and oxidative stress in human type II alveolar epithelial cells. In this study, A549 cells were transfected with Nrf2 siRNA and overexpression vectors for 6 h before being induced by TNF-α for 24 h. TNF-α upregulated the expression of NOX1 and Nrf2 in A549 cells. Furthermore, overexpression of Nrf2 could reduce TNF-α-induced NF-κB mRNA and protein expression after transfection with the Nrf2 siRNA vector, and the levels of IL-6, IL-8, ROS, and malondialdehyde (MDA) in TNF-α-induced A549 cells increased, while the level of total antioxidation capability (T-AOC) decreased. On the other hand, the overexpression of Nrf2 decreased the levels of IL-6, IL-8, ROS, and MDA, while increasing T-AOC. The mRNA and protein levels of NOX1 were dramatically increased by TNF-α, while those changes were notably suppressed by Nrf2 overexpression. Further studies demonstrated that Nrf2 suppressed NOX1 transcription by binding to the -1199 to -1189 bp (ATTACACAGCA) region of the NOX1 promoter in TNF-α-stimulated A549 cells. Our study suggests that Nrf2 may bind to and regulate NOX1 expression to antagonize TNF-α-induced inflammatory reaction and oxidative stress in A549 cells.
Collapse
|
13
|
Fu H, Zhu H. Geniposidic acid protects lipopolysaccharide-induced acute lung injury via the TLR4/MyD88 signaling pathway in vitro and in vivo. Immunopharmacol Immunotoxicol 2022; 44:984-992. [PMID: 35770920 DOI: 10.1080/08923973.2022.2096465] [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: 12/13/2022]
Abstract
BACKGROUND Acute lung injury (ALI) is a common respiratory disease and is a serious threat to human health due to the lack of effective treatment. Geniposidic acid (GPA) is an iridoid glucoside extracted from Gardeniae jasminoides Ellis and can treat inflammation-related diseases. This study aimed to investigate the regulatory functions of GPA on lipopolysaccharide (LPS)-induced ALI and its potential mechanism, providing effective strategies for the clinical treatment of ALI. METHODS ALI models were constructed by LPS in Sprague-Dawley rats and pulmonary epithelial cells. The function of GPA was investigated by hematoxylin-eosin staining, lung function assessment, Western blot, Masson staining, and Sirius Red staining, quantitative real-time PCR, enzyme-linked immunosorbent assay, cell counting kit-8 assay, apoptosis analysis, and immunofluorescence assays. RESULTS Functionally, GPA increased survival, relieved pulmonary epithelial function in response to LPS, repressed pulmonary fibrosis and inflammation caused by ALI in vivo; GPA also repressed pulmonary epithelial cell injury and inflammation induced by LPS in vitro. Mechanistically, GPA decreased the protein levels of TLR4 and MyD88 and accelerated the nuclear export of p65, suggesting that GPA repressed the activation of p65. CONCLUSION GPA protected LPS-induced ALI through the TLR4/MyD88 signaling pathway.
Collapse
Affiliation(s)
- Hui Fu
- Department of Pediatrics, Changzhou Second People's Hospital, Changzhou, China
| | - Hui Zhu
- Department of Pediatrics, Nantong Hospital of Traditional Chinese Medicine, Nantong, China
| |
Collapse
|
14
|
Hong H, Lou S, Zheng F, Gao H, Wang N, Tian S, Huang G, Zhao H. Hydnocarpin D attenuates lipopolysaccharide-induced acute lung injury via MAPK/NF-κB and Keap1/Nrf2/HO-1 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 101:154143. [PMID: 35537248 DOI: 10.1016/j.phymed.2022.154143] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/22/2022] [Accepted: 05/01/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Acute lung injury (ALI) is a complex pulmonary destructive disease with limited therapeutic approaches. Hydnocarpin D (HD) is a flavonolignan isolated from Hydnocarpus wightiana which possesses antioxidant and anti-inflammatory properties. However, whether HD has beneficial effects on ALI as well as its underlying mechanism remains to be elucidated. PURPOSE This study evaluated the protective effect of HD in ALI and the underlying molecular mechanisms. METHODS In vivo, the role of HD on lipopolysaccharide (LPS)-induced ALI in mice was tested by determination of neutrophil infiltration, levels of inflammatory cytokines, lung histology and edema, vascular and alveolar barrier disruption. In vitro, murine macrophage RAW 264.7 cells were used to investigate the molecular mechanisms RESULTS: Administration of HD protected mice against LPS-induced ALI, including ameliorating the histological alterations in the lung tissues, and decreasing lung edema, protein content of bronchoalveolar lavage fluid, infiltration of inflammatory cell and secretion of cytokines. Moreover, HD blocked the phosphorylation of TLR-4, NF-κB, and ERK in LPS-induced lung injury. In vitro, HD inhibited LPS-induced oxidative stress and inflammation in RAW 264.7 cells, which largely depend upon the upregulation of antioxidant defensive Nrf2 pathway, thereby suppressing LPS-activated proinflammatory mediator secretion, NLRP3 inflammasome, and MAPK/NF-κB signaling pathway. CONCLUSION HD attenuates oxidative stress and inflammation against LPS-induced ALI via MAPK/NF-κB and Keap1/Nrf2/HO-1 pathway, and is a promising novel therapeutic candidate for ALI.
Collapse
Affiliation(s)
- Huanwu Hong
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Siyue Lou
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Fanli Zheng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Hang Gao
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Nina Wang
- College of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan 243002, China
| | - Shasha Tian
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Guozheng Huang
- College of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan 243002, China.
| | - Huajun Zhao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
15
|
Wang Y, Yuan Y, Wang W, He Y, Zhong H, Zhou X, Chen Y, Cai XJ, Liu LQ. Mechanisms underlying the therapeutic effects of Qingfeiyin in treating acute lung injury based on GEO datasets, network pharmacology and molecular docking. Comput Biol Med 2022; 145:105454. [DOI: 10.1016/j.compbiomed.2022.105454] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/09/2022] [Accepted: 03/23/2022] [Indexed: 12/11/2022]
|
16
|
Zhang Y, Zhang J, Fu Z. Molecular hydrogen is a potential protective agent in the management of acute lung injury. Mol Med 2022; 28:27. [PMID: 35240982 PMCID: PMC8892414 DOI: 10.1186/s10020-022-00455-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 02/14/2022] [Indexed: 11/21/2022] Open
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome, which is a more severe form of ALI, are life-threatening clinical syndromes observed in critically ill patients. Treatment methods to alleviate the pathogenesis of ALI have improved to a great extent at present. Although the efficacy of these therapies is limited, their relevance has increased remarkably with the ongoing pandemic caused by the novel coronavirus disease 2019 (COVID-19), which causes severe respiratory distress syndrome. Several studies have demonstrated the preventive and therapeutic effects of molecular hydrogen in the various diseases. The biological effects of molecular hydrogen mainly involve anti-inflammation, antioxidation, and autophagy and cell death modulation. This review focuses on the potential therapeutic effects of molecular hydrogen on ALI and its underlying mechanisms and aims to provide a theoretical basis for the clinical treatment of ALI and COVID-19.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Jin Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Zhiling Fu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.
| |
Collapse
|
17
|
Ge J, Yang H, Zeng Y, Liu Y. Protective effects of wogonin on lipopolysaccharide-induced inflammation and apoptosis of lung epithelial cells and its possible mechanisms. Biomed Eng Online 2021; 20:125. [PMID: 34906140 PMCID: PMC8670054 DOI: 10.1186/s12938-021-00965-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 11/29/2021] [Indexed: 01/02/2023] Open
Abstract
Background Wogonin (5, 7-dihydroxy-8-methoxyflavone) is a natural di-hydroxyl flavonoid extracted from the root of Scutellaria baicalensis Georgi. This paper was intended to investigate the mechanism of action of wogonin in alleviating the inflammation and apoptosis in acute lung injury (ALI). Materials and methods Lipopolysaccharide (LPS) was used to establish the in vitro model of ALI. After wogonin treatment, the cell viability and apoptosis of LPS-induced A549 cells were, respectively, measured by CCK-8, TUNEL assays and acridine orange/ethidium bromide dual staining, while the contents of inflammatory cytokines and oxidative stress markers were estimated by RT-qPCR, ELISA assay, western blot analysis and commercial kits. Western blot was also conducted to assess the expression of proteins involved. Subsequently, the effect of wogonin on the sirtuin 1 (SIRT1)-mediated high-mobility group box 1 protein (HMGB1) deacetylation was investigated. SIRT1 inhibitor EX527 was used to evaluate the regulatory effects of wogonin on SIRT1-mediated HMGB1 deacetylation in A549 cells under LPS stimulation. Results LPS induced inflammation, oxidative stress and apoptosis of A549 cells, which was abolished by wogonin. It was also found that wogonin promoted the HMGB1 deacetylation, accompanied by upregulated SIRT1 expression. However, SIRT1 inhibitor EX527 partially reversed the protective effects of wogonin on the inflammation and apoptosis of LPS-induced A549 cells. Conclusion Wogonin alleviated the inflammation and apoptosis in LPS-induced A549 cells by SIRT1-mediated HMGB1 deacetylation, which might represent the identification of a novel mechanism by which wogonin exerts protective effects on ALI and provide ideas for the application of wogonin to ALI treatment.
Collapse
Affiliation(s)
- Jinlin Ge
- Department of Pulmonary and Critical Care Medicine, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Wenzhou, 325000, Zhejiang Province, China
| | - Huanhuan Yang
- Department of Pulmonary and Critical Care Medicine, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Wenzhou, 325000, Zhejiang Province, China
| | - Yufeng Zeng
- Department of Pulmonary and Critical Care Medicine, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Wenzhou, 325000, Zhejiang Province, China
| | - Yunjie Liu
- Department of Respiratory and Critical Care Medicine, The Second People's Hospital of Nantong, 298 Xinhua Road, Chongchuan District, Nantong, 226002, Jiangsu, China.
| |
Collapse
|
18
|
Montanucci P, Pescara T, Greco A, Francisci D, Basta G, Calafiore R. Microencapsulated Wharton Jelly-derived adult mesenchymal stem cells as a potential new therapeutic tool for patients with COVID-19 disease: an in vitro study. AMERICAN JOURNAL OF STEM CELLS 2021; 10:36-52. [PMID: 34552816 PMCID: PMC8449139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 08/07/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND The recent newly appeared Coronavirus disease (COVID-19), caused by an enveloped RNA virus named "severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)", is associated with severe respiratory morbidity and mortality. Recent studies have shown that lymphopenia and a cytokine mass release represent important pathogenic features, with clinical evidence of dyspnea and hypoxemia, often leading to acute respiratory distress syndrome (ARDS), in severely ill patients, with a high death toll. Currently, stem cells are actively being investigated for their potential use in many "untreatable" diseases. In this regard and in particular, Mesenchymal Stem Cells (MSC), due to their intrinsic features, including either ability to impact on regulation of the immune system, or association with both anti-viral and anti-inflammatory properties, or potential for differentiation into several cell lineages, have become a promising tool for cell and molecular-based therapies. On this background, we wished to explore whether human umbilical cord-derived mesenchymal stem cells (hUCMS) would represent a potential viable therapeutic approach for the management of critically ill COVID19 patients. METHODS We tested the hUCMS effects on peripheral blood mononuclear cell (PBMCs) retrieved from patients with COVID19 (Ethical Committee CEAS Umbria, Italy CER N°3658/20 7, May, 2020), both as free cell monolayers and after envelopment in sodium alginate microcapsules. Both cell systems, after priming with IFN-γ, proved able to produce several immunomodulatory molecules such as IDO1 and HLAG5, although only the microencapsulated hUCMS were associated with massive and dose-dependent production of these factors. RESULTS The microencapsulated hUCMS improved allo-suppression in mixed lymphocytes reactions (MLRs), while also blunting T helper 1 and T helper 17 responses, that are involved with the cytokine storm and greatly contribute to the patient death. Moreover, we observed that both free and microencapsulated hUCMS permitted 5 days survival of in vitro culture maintained PBMCs extracted from very ill patients. CONCLUSION We have provided evidence that microencapsulated hUCMS in vitro, seem to represent a powerful tool to impact on several immune pathways, clearly deranged in COVID19 patients. Further study is necessary to begin in vivo assessment of this experimental system, upon determining both, the most appropriate time of the disease onset for intervention, and cell dosage/patient of our experimental product.
Collapse
Affiliation(s)
- Pia Montanucci
- Division of Internal Medicine and Endocrine and Metabolic Sciences (MISEM), Laboratory for Endocrine Cell Transplants and Biohybrid Organs, Department of Medicine and Surgery, University of PerugiaPiazzale Gambuli I-06132, Perugia, Italy
| | - Teresa Pescara
- Division of Internal Medicine and Endocrine and Metabolic Sciences (MISEM), Laboratory for Endocrine Cell Transplants and Biohybrid Organs, Department of Medicine and Surgery, University of PerugiaPiazzale Gambuli I-06132, Perugia, Italy
| | - Alessia Greco
- Division of Internal Medicine and Endocrine and Metabolic Sciences (MISEM), Laboratory for Endocrine Cell Transplants and Biohybrid Organs, Department of Medicine and Surgery, University of PerugiaPiazzale Gambuli I-06132, Perugia, Italy
| | - Daniela Francisci
- Division of Infectious Diseases, Department of Medicine and Surgery, University of PerugiaPiazzale Gambuli I-06132, Perugia, Italy
| | - Giuseppe Basta
- Division of Internal Medicine and Endocrine and Metabolic Sciences (MISEM), Laboratory for Endocrine Cell Transplants and Biohybrid Organs, Department of Medicine and Surgery, University of PerugiaPiazzale Gambuli I-06132, Perugia, Italy
| | - Riccardo Calafiore
- Division of Internal Medicine and Endocrine and Metabolic Sciences (MISEM), Laboratory for Endocrine Cell Transplants and Biohybrid Organs, Department of Medicine and Surgery, University of PerugiaPiazzale Gambuli I-06132, Perugia, Italy
| |
Collapse
|
19
|
Fengyun W, LiXin Z, Xinhua Q, Bin F. Mesenchymal Stromal Cells Attenuate Infection-Induced Acute Respiratory Distress Syndrome in Animal Experiments: A Meta-Analysis. Cell Transplant 2021; 29:963689720969186. [PMID: 33164559 PMCID: PMC7784610 DOI: 10.1177/0963689720969186] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stromal cell (MSC) therapy is a potential therapy for treating acute lung injury (ALI) or acute respiratory distress syndrome (ARDS), which was widely studied in the last decade. The purpose of our meta-analysis was to investigate the efficacy of MSCs for simulated infection-induced ALI/ARDS in animal trials. PubMed and EMBASE were searched to screen relevant preclinical trials with a prespecified search strategy. 57 studies met the inclusion criteria and were included in our study. Our meta-analysis showed that MSCs can reduce the lung injury score of ALI caused by lipopolysaccharide or bacteria (standardized mean difference (SMD) = −2.97, 95% CI [−3.64 to −2.30], P < 0.00001) and improve the animals’ survival (odds ratio = 3.64, 95% CI [2.55 to 5.19], P < 0.00001). Our study discovered that MSCs can reduce the wet weight to dry weight ratio of the lung (SMD = −2.58, 95% CI [−3.24 to −1.91], P < 0.00001). The proportion of the alveolar sac in the MSC group was higher than that in the control group (SMD = 1.68, 95% CI [1.22 to 2.13], P < 0.00001). Moreover, our study detected that MSCs can downregulate the levels of proinflammatory factors such as interleukin (IL)-1β, IL-6, and tumor necrosis factor-α in the lung and it can upregulate the level of anti-inflammatory factor IL-10. MSCs were also found to reduce the level of neutrophils and total protein in bronchoalveolar lavage fluid, decrease myeloperoxidase (MPO) activity in the lung, and improve lung compliance. MSC therapy may be a promising treatment for ALI/ARDS since it may mitigate the severity of lung injury, modulate the immune balance, and ameliorate the permeability of lung vessels in ALI/ARDS, thus facilitating lung regeneration and repair.
Collapse
Affiliation(s)
- Wang Fengyun
- Department of Critical Care Medicine, 66278The First People's Hospital of Foshan, Foshan, China
| | - Zhou LiXin
- Department of Critical Care Medicine, 66278The First People's Hospital of Foshan, Foshan, China
| | - Qiang Xinhua
- Department of Critical Care Medicine, 66278The First People's Hospital of Foshan, Foshan, China
| | - Fang Bin
- Department of Critical Care Medicine, 66278The First People's Hospital of Foshan, Foshan, China
| |
Collapse
|
20
|
Therapeutic Applications of Stem Cells and Extracellular Vesicles in Emergency Care: Futuristic Perspectives. Stem Cell Rev Rep 2021; 17:390-410. [PMID: 32839921 PMCID: PMC7444453 DOI: 10.1007/s12015-020-10029-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Regenerative medicine (RM) is an interdisciplinary field that aims to repair, replace or regenerate damaged or missing tissue or organs to function as close as possible to its physiological architecture and functions. Stem cells, which are undifferentiated cells retaining self-renewal potential, excessive proliferation and differentiation capacity into offspring or daughter cells that form different lineage cells of an organism, are considered as an important part of the RM approaches. They have been widely investigated in preclinical and clinical studies for therapeutic purposes. Extracellular vesicles (EVs) are the vital mediators that regulate the therapeutic effects of stem cells. Besides, they carry various types of cargo between cells which make them a significant contributor of intercellular communication. Given their role in physiological and pathological conditions in living cells, EVs are considered as a new therapeutic alternative solution for a variety of diseases in which there is a high unmet clinical need. This review aims to summarize and identify therapeutic potential of stem cells and EVs in diseases requiring acute emergency care such as trauma, heart diseases, stroke, acute respiratory distress syndrome and burn injury. Diseases that affect militaries or societies including acute radiation syndrome, sepsis and viral pandemics such as novel coronavirus disease 2019 are also discussed. Additionally, featuring and problematic issues that hamper clinical translation of stem cells and EVs are debated in a comparative manner with a futuristic perspective. Graphical Abstract.
Collapse
|
21
|
Majolo F, da Silva GL, Vieira L, Timmers LFSM, Laufer S, Goettert MI. Review of Trials Currently Testing Stem Cells for Treatment of Respiratory Diseases: Facts Known to Date and Possible Applications to COVID-19. Stem Cell Rev Rep 2021; 17:44-55. [PMID: 32827081 PMCID: PMC7442550 DOI: 10.1007/s12015-020-10033-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Therapeutic clinical and preclinical studies using cultured cells are on the rise, especially now that the World Health Organization (WHO) declared coronavirus disease 2019 (COVID-19) a "public health emergency of international concern", in January, 2020. Thus, this study aims to review the outcomes of ongoing clinical studies on stem cells in Severe Acute Respiratory Syndrome (SARS), Acute Respiratory Distress Syndrome (ARDS), and Middle East Respiratory Syndrome (MERS). The results will be associated with possible applications to COVID-19. Only three clinical trials related to stem cells are considered complete, whereby two are in Phase 1 and one is in Phase 2. Basically, the ongoing studies on coronavirus are using mesenchymal stem cells (MSCs) derived from bone marrow or the umbilical cord to demonstrate their feasibility, safety, and tolerability. The studies not related to coronavirus are all in ARDS conditions; four of them are in Phase 1 and three in Phase 2. With the COVID-19 boom, many clinical trials are being carried out using different sources with an emphasis on MSC-based therapy used to inhibit inflammation. One of the biggest challenges in the current treatment of COVID-19 is the cytokine storm, however MSCs can prevent or mitigate this cytokine storm through their immunomodulatory capacity. We look forward to the results of the ongoing clinical trials to find a treatment for the disease. Researchers around the world are joining forces to help fight COVID-19. Stem cells used in the current clinical studies are a new therapeutic promise for COVID-19 where pharmacological treatments seem insufficient.Graphical Abstract.
Collapse
Affiliation(s)
- Fernanda Majolo
- Post-graduate Program in Biotechnology, Universidade do Vale do Taquari - Univates, Av. Avelino Talini, 171, 95914-014, Lajeado, Rio Grande do Sul, Brazil
| | - Guilherme Liberato da Silva
- Medical Sciences Center, Universidade do Vale do Taquari - Univates, Lajeado, Rio Grande do Sul, 95914-014, Brazil
| | - Lucas Vieira
- Medical Sciences Center, Universidade do Vale do Taquari - Univates, Lajeado, Rio Grande do Sul, 95914-014, Brazil
| | - Luís Fernando Saraiva Macedo Timmers
- Post-graduate Program in Biotechnology, Universidade do Vale do Taquari - Univates, Av. Avelino Talini, 171, 95914-014, Lajeado, Rio Grande do Sul, Brazil
| | - Stefan Laufer
- Medicinal Chemistry, University of Tuebingen, D-72076, Tubingen, Germany
| | - Márcia Inês Goettert
- Post-graduate Program in Biotechnology, Universidade do Vale do Taquari - Univates, Av. Avelino Talini, 171, 95914-014, Lajeado, Rio Grande do Sul, Brazil.
| |
Collapse
|
22
|
Wang Y, Li H, Li X, Su X, Xiao H, Yang J. Hypoxic Preconditioning of Human Umbilical Cord Mesenchymal Stem Cells Is an Effective Strategy for Treating Acute Lung Injury. Stem Cells Dev 2021; 30:128-134. [PMID: 33349130 DOI: 10.1089/scd.2020.0174] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS)/acute lung injury (ALI) is a severe clinical respiratory failure disorder associated with chronic pathology and disability and has a mortality rate of 40%-60%. However, the pathogenesis of ARDS/ALI remains unclear, and existing therapeutic options are insufficient for addressing the severity of the disease. Mesenchymal stem cells (MSCs) play an important role in the prevention and treatment of ALI, especially acute alveolar epithelial injury. However, the low survival rate of transplanted MSCs reduces their effectiveness. When human umbilical cord MSCs (hUC-MSCs) are transplanted directly, only a minority of cells migrate toward damaged tissues. Moreover, their maintenance time is short, leading to unsatisfactory therapeutic results. A moderate hypoxic environment can promote the proliferation of MSCs, inhibit apoptosis, and facilitate migration and chemotaxis. In summary, hypoxic culturing before transplantation improves the effectiveness of hUC-MSCs in treating ARDS/ALI and promises to provide novel diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Yujuan Wang
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Han Li
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Xue Li
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Xin Su
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Han Xiao
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Junling Yang
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
23
|
Abstract
The COVID-19 pandemic has led to a major setback in both the health and economic sectors across the globe. The scale of the problem is enormous because we still do not have any specific anti-SARS-CoV-2 antiviral agent or vaccine. The human immune system has never been exposed to this novel virus, so the viral interactions with the human immune system are completely naive. New approaches are being studied at various levels, including animal in vitro models and human-based studies, to contain the COVID-19 pandemic as soon as possible. Many drugs are being tested for repurposing, but so far only remdesivir has shown some positive benefits based on preliminary reports, but these results also need further confirmation via ongoing trials. Otherwise, no other agents have shown an impactful response against COVID-19. Recently, research exploring the therapeutic application of mesenchymal stem cells (MSCs) in critically ill patients suffering from COVID-19 has gained momentum. The patients belonging to this subset are most likely beyond the point where they could benefit from an antiviral therapy because most of their illness at this stage of disease is driven by inflammatory (over)response of the immune system. In this review, we discuss the potential of MSCs as a therapeutic option for patients with COVID-19, based on the encouraging results from the preliminary data showing improved outcomes in the progression of COVID-19 disease.
Collapse
Affiliation(s)
- Kamal Kant Sahu
- Department of Hematology and Oncology, Department of Internal Medicine, Saint Vincent Hospital, Worcester, Massachusetts
| | - Ahmad Daniyal Siddiqui
- Department of Hematology and Oncology, Department of Internal Medicine, Saint Vincent Hospital, Worcester, Massachusetts
| | - Jan Cerny
- Division of Hematology and Oncology, Department of Medicine, UMass Memorial Health Care, University of Massachusetts Medical School, Worcester, Massachusetts
| |
Collapse
|
24
|
Wei F, Kong D, Li T, Li A, Tan Y, Fang J, Zhuang X, Lai C, Xu W, Dong H, Ma C, Hong K, Cui Y, Tang S, Yu F, Zheng C. Efficacy and safety of umbilical cord mesenchymal stem cells for the treatment of patients with COVID-19. Clinics (Sao Paulo) 2021; 76:e2604. [PMID: 34008772 PMCID: PMC8101688 DOI: 10.6061/clinics/2021/e2604] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/29/2021] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES The coronavirus disease (COVID-19) outbreak has catastrophically threatened public health worldwide and presented great challenges for clinicians. To date, no specific drugs are available against severe acute respiratory syndrome coronavirus 2. Mesenchymal stem cells (MSCs) appear to be a promising cell therapy owing to their potent modulatory effects on reducing and healing inflammation-induced lung and other tissue injuries. The present pilot study aimed to explore the therapeutic potential and safety of MSCs isolated from healthy cord tissues in the treatment of patients with COVID-19. METHODS Twelve patients with COVID-19 treated with MSCs plus conventional therapy and 13 treated with conventional therapy alone (control) were included. The efficacy of MSC infusion was evaluated by changes in oxygenation index, clinical chemistry and hematology tests, immunoglobulin (Ig) levels, and pulmonary computerized tomography (CT) imaging. The safety of MSC infusion was evaluated based on the occurrence of allergic reactions and serious adverse events. RESULTS The MSC-treated group demonstrated significantly improved oxygenation index. The area of pulmonary inflammation decreased significantly, and the CT number in the inflammatory area tended to be restored. Decreased IgM levels were also observed after MSC therapy. Laboratory biomarker levels at baseline and after therapy showed no significant changes in either the MSC-treated or control group. CONCLUSION Intravenous infusion of MSCs in patients with COVID-19 was effective and well tolerated. Further studies involving a large cohort or randomized controlled trials are warranted.
Collapse
Affiliation(s)
- Fengtao Wei
- Department of Cardiology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Corresponding authors. E-mails: / /
| | - Dexiao Kong
- Department of Hematology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tao Li
- Department of Infectious Disease and Hepatology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ai Li
- Department of Hematology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yi Tan
- Shandong Qilu Cell Therapy Engineering Tecnology Co. Ltd, Jinan, Shandong Province, China
| | - Jinfeng Fang
- Department of Hematology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xianghua Zhuang
- Department of Endocrinology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chao Lai
- Department of Neurology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Weihua Xu
- Department of Gastroenterology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hong Dong
- Nursing Department, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chengen Ma
- Department of Critical Care Medicine, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ke Hong
- Yinfeng Gene Technology Co. Ltd, Jinan, Shandong Province, China
| | - Yuqin Cui
- Shandong Yinfeng Institute of Life Science, Jinan, Shandong Province, China
| | - Shengbin Tang
- Hunan Sheng Bao Biological Technology Co. Ltd, GaoXin District, ChangSha, China
| | - Fenggang Yu
- Shandong Yinfeng Institute of Life Science, Jinan, Shandong Province, China
- Institute for Advanced Interdisciplinary Research, University of Jinan, Jinan, 250022, China
- *Corresponding authors. E-mails: / /
| | - Chengyun Zheng
- Department of Hematology, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Corresponding authors. E-mails: / /
| |
Collapse
|
25
|
Sadeghi S, Soudi S, Shafiee A, Hashemi SM. Mesenchymal stem cell therapies for COVID-19: Current status and mechanism of action. Life Sci 2020; 262:118493. [PMID: 32979360 PMCID: PMC7510562 DOI: 10.1016/j.lfs.2020.118493] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/19/2020] [Accepted: 09/20/2020] [Indexed: 12/12/2022]
Abstract
The outbreak of COVID-19 in December 2019, has become an urgent and serious public health emergency. At present, there is no effective treatment or vaccine for COVID-19. Therefore, there is a crucial unmet need to develop a safe and effective treatment for COVID-19 patients. Mesenchymal stem cells (MSCs) are widely used in basic science and in a variety of clinical trials. MSCs are able to engraft to the damaged tissues after transplantation and promote tissue regeneration, besides MSCs able to secrete immunomodulatory factors that suppress the cytokine storms. Moreover, the contribution of MSCs to prevent cell death and inhibit tissue fibrosis is well established. In the current review article, the potential mechanisms by which MSCs contribute to the treatment of COVID-19 patients are highlighted. Also, current trials that evaluated the potential of MSC-based treatments for COVID-19 are briefly reviewed.
Collapse
Affiliation(s)
- Somaye Sadeghi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Soudi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Abbas Shafiee
- UQ Diamantina Institute, Translational Research Institute, The University of Queensland, Brisbane, Queensland, Australia.
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
26
|
Lian J, Lin J, Zakaria N, Yahaya BH. Acute Lung Injury: Disease Modelling and the Therapeutic Potential of Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1298:149-166. [PMID: 32424492 DOI: 10.1007/5584_2020_538] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Acute lung injury (ALI) is a severe clinical condition with high morbidity and mortality that usually results in the development of multiple organ dysfunction. The complex pathophysiology of ALI seems to provide a wide range of targets that offer numerous therapeutic options. However, despite extensive studies of ALI pathophysiology and treatment, no effective pharmacotherapy is available. Increasing evidence from both preclinical and clinical studies supports the preventive and therapeutic effects of mesenchymal stem cells (MSCs) for treating ALI. As cell-based therapy poses the risk of occlusion in microvasculature or unregulated growth, MSC-derived extracellular vesicles (MSC-EVs) have been extensively studied as a new therapeutic strategy for non-cell based therapy. It is widely accepted that the therapeutic properties of MSCs are derived from soluble factors with paracrine or endocrine effects, and EVs are among the most important paracrine or endocrine vehicles that can deliver various soluble factors with a similar phenotype as the parent cell. Therapeutic effects of MSCs have been reported for various delivery approaches, diverse doses, multiple origins, and different times of administration, and MSC-EVs treatment may include but is not limited to these choices. The mechanisms by which MSCs and MSC-EVs may contribute to ALI treatment remain elusive and need further exploration. This review provides an overview of preclinical studies that support the application of MSC-EVs for treating ALI, and it discusses emerging opportunities and their associated challenges.
Collapse
Affiliation(s)
- Jie Lian
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), Universiti Sains Malaysia, SAINS@Bertam, Penang, Malaysia.,Stem Cell and Biotherapy Technology Research Center of Henan Province, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Juntang Lin
- Stem Cell and Biotherapy Technology Research Center of Henan Province, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Norashikin Zakaria
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), Universiti Sains Malaysia, SAINS@Bertam, Penang, Malaysia
| | - Badrul Hisham Yahaya
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), Universiti Sains Malaysia, SAINS@Bertam, Penang, Malaysia.
| |
Collapse
|
27
|
Dubus M, Varin-Simon J, Prada P, Scomazzon L, Reffuveille F, Alem H, Boulmedais F, Mauprivez C, Rammal H, Kerdjoudj H. Biopolymers-calcium phosphate antibacterial coating reduces the pathogenicity of internalized bacteria by mesenchymal stromal cells. Biomater Sci 2020; 8:5763-5773. [PMID: 32945302 DOI: 10.1039/d0bm00962h] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A multifunctional material system that kills bacteria and drives bone healing is urgently sought to improve bone prosthesis. Herein, the osteoinductive coating made of calcium phosphate/chitosan/hyaluronic acid, named Hybrid, was proposed as an antibacterial substrate for stromal cell adhesion. This Hybrid coating possesses a contact-killing effect reducing by 90% the viability of Gram-positive Staphylococcus aureus (S. aureus) and Gram-negative Pseudomonas aeruginosa (P. aeruginosa) strains after 48 h of contact. In addition to the production of immunomodulatory mediators, Wharton's jelly (WJ-SCs), dental pulp (DPSCs) and bone marrow (BM-MSCs) derived stromal cells were able to release antibacterial and antibiofilm agents effective against S. aureus and P. aeruginosa strains, respectively. Studying the effect of the Hybrid coating on the internalization of S. aureus by the stromal cells, in acute-mimicking bone infection, highlighted an increase in the bacteria internalization by DPSCs and BM-MSCs when cultured on the Hybrid coating versus uncoated glass. Despite the internalization, Hybrid coating showed a beneficial effect by reducing the pathogenicity of the internalized bacteria. The formation of biofilm was reduced by at least 50% in comparison to internalized bacteria by stromal cells on uncoated glass. This work opens the route for the development of innovative antibacterial coatings by taking into account the internalization of bacteria by stromal cells.
Collapse
Affiliation(s)
- Marie Dubus
- Université de Reims Champagne Ardenne, EA 4691, Biomatériaux et Inflammation en Site Osseux (BIOS), Reims, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Yu F, Jia R, Tang Y, Liu J, Wei B. SARS-CoV-2 infection and stem cells: Interaction and intervention. Stem Cell Res 2020; 46:101859. [PMID: 32570174 PMCID: PMC7263221 DOI: 10.1016/j.scr.2020.101859] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/22/2020] [Accepted: 05/25/2020] [Indexed: 02/08/2023] Open
Abstract
The emergence of the novel severe acute respiratory coronavirus 2 (SARS-CoV-2) in China and its rapid national and international spread have created a global health emergency. The resemblance with SARS-CoV in spike protein suggests that SARS-CoV-2 employs spike-driven entry into angiotensin-converting enzyme 2 (ACE2)-expressing cells. From a stem cell perspective, this review focuses on the possible involvement of ACE2+ stem/progenitor cells from both the upper and lower respiratory tracts in coronavirus infection. Viral infection-associated acute respiratory distress syndrome and acute lung injury occur because of dysregulation of the immune response. Mesenchymal stem cells appear to be a promising cell therapy given that they favorably modulate the immune response to reduce lung injury. The use of exogenous stem cells may lead to lung repair. Therefore, intervention by transplantation of exogenous stem cells may be required to replace, repair, remodel, and regenerate lung tissue in survivors infected with coronavirus. Ultimately, vaccines, natural killer cells and induced-pluripotent stem cell-derived virus-specific cytotoxic T lymphocytes may offer off-the-shelf therapeutics for preventing coronavirus reemergence.
Collapse
Affiliation(s)
- Fenggang Yu
- Institute of Life Science, Yingfeng Bilogical Group, Jinan, Shandong Province, China; Institute for Advanced Interdisciplinary Researc (iAIR), University of Jinan, Jinan 250022, China.
| | - Rufu Jia
- Cangzhou Central Hospital, Cangzhou, Hebei Province, China
| | - Yongyong Tang
- Yinfeng Dingcheng Bioengineering and Technology Ltd, Beijing, China
| | - Jin Liu
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China; Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Benjie Wei
- Institute of Life Science, Yingfeng Bilogical Group, Jinan, Shandong Province, China
| |
Collapse
|
29
|
Jin C, Zhou F, Zhang L, Shen J. Overexpression of heat shock protein 70 enhanced mesenchymal stem cell treatment efficacy in phosgene-induced acute lung injury. J Biochem Mol Toxicol 2020; 34:e22515. [PMID: 32394537 DOI: 10.1002/jbt.22515] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 04/22/2020] [Indexed: 12/19/2022]
Abstract
In our previous study, we have confirmed that in phosgene-induced acute lung injury (ALI) rats, mesenchymal stem cells (MSCs) can treat the disease. Moreover, heat shock protein 70 (Hsp70) can be used as a protective protein, and Hsp70 upregulated drastically when exposed to stressful conditions. We aimed to assess that MSCs overexpressed Hsp70 could enhance the capacity of MSCs and have a good therapeutic effect on phosgene-induced ALI. We transduced MSCs with Hsp70 and then we tested the function of the transduced MSCs. Sprague Dawley rats inhaled phosgene in a closed container for 5 minutes. The transduced MSCs and MSCs were administered via the trachea immediately. Rats in each group were killed at 6, 24, and 48 hours after exposure. Compared to MSCs, MSCs overexpressed Hsp70 enhanced MSCs viability, antiapoptotic ability, and migration ability, and these effects disappeared when using the phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) pathway inhibitor. Furthermore, the results of pathological alterations improved. The lung wet-to-dry ratio declined. The lung injury index total protein content and total cells in bronchoalveolar lavage fluid (BALF) also declined. The level of tumor necrosis factor α declined and the level of interleukin-10 improved in BALF and serum. MSCs overexpressed Hsp70 can enhance the capacity and efficacy of MSCs in the treatment of phosgene-induced ALI and may be mediated through the PI3k/AKT signaling pathway. This article introduces a new approach to stem cell therapy for improving the efficacy of phosgene-induced ALI.
Collapse
Affiliation(s)
- Chaoyuan Jin
- Center of Emergency and Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, China.,Medical Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China.,Medical Research Center for Chemical Injury, Emergency and Critical Care of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China
| | - Fangqing Zhou
- Center of Emergency and Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, China.,Medical Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China.,Medical Research Center for Chemical Injury, Emergency and Critical Care of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China
| | - Lin Zhang
- Center of Emergency and Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, China.,Medical Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China.,Medical Research Center for Chemical Injury, Emergency and Critical Care of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China
| | - Jie Shen
- Center of Emergency and Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, China.,Medical Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China.,Medical Research Center for Chemical Injury, Emergency and Critical Care of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
30
|
Sadeghian Chaleshtori S, Mokhber Dezfouli MR, Jabbari Fakhr M. Mesenchymal stem/stromal cells: the therapeutic effects in animal models of acute pulmonary diseases. Respir Res 2020; 21:110. [PMID: 32393278 PMCID: PMC7213547 DOI: 10.1186/s12931-020-01373-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 04/23/2020] [Indexed: 02/06/2023] Open
Abstract
The pulmonary diseases are one of the most important causes of death in the world. The successful therapies in the field of lung diseases are very limited and the medical treatments available are ineffective in many of the lung diseases. Many studies have evaluated the new therapies in the acute pulmonary diseases, and the transplantation of mesenchymal stem/stromal cells (MSCs), which is a branch of cell therapy, has a special place among the new medical techniques. The MSCs are present throughout the body and are thought to play a role in tissue regeneration and inflammation control. In the event of injury, the local MSCs traverse the shortest possible distance from the tissue or blood vessels to reach the affected site. But, there are few undifferentiated cells in the tissues. The exogenous MSCs are used to immunity modify or regenerative treatments in preclinical models of acute pulmonary diseases. Several studies have shown the positive effects of MSCs replacement in the acute lung disorders. The effection mechanism of the MSCs include the differentiation ability and the secretion of paracrine agents such as the anti-inflammatory mediators. Many studies suggest that this treatment method is safe and is probably to be widely used in future clinical trials. This review will describe the therapeutic effects of the MSCs in the experimental models of the acute pulmonary diseases for use as a method of treatment in clinical trials in future.
Collapse
Affiliation(s)
- Sirous Sadeghian Chaleshtori
- Department of Internal Medicine, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.,Institute of Biomedical Research, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mohammad Reza Mokhber Dezfouli
- Department of Internal Medicine, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran. .,Institute of Biomedical Research, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| | - Massoumeh Jabbari Fakhr
- Institute of Biomedical Research, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.,Department of Tissue Engineering, Faculty of Medicine, Qom University of Medical Sciences, Qom, Iran
| |
Collapse
|
31
|
Thong T, Forté CA, Hill EM, Colacino JA. Environmental exposures, stem cells, and cancer. Pharmacol Ther 2019; 204:107398. [PMID: 31376432 PMCID: PMC6881547 DOI: 10.1016/j.pharmthera.2019.107398] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 07/29/2019] [Indexed: 12/19/2022]
Abstract
An estimated 70-90% of all cancers are linked to exposure to environmental risk factors. In parallel, the number of stem cells in a tissue has been shown to be a strong predictor of risk of developing cancer in that tissue. Tumors themselves are characterized by an acquisition of "stem cell" characteristics, and a growing body of evidence points to tumors themselves being sustained and propagated by a stem cell-like population. Here, we review our understanding of the interplay between environmental exposures, stem cell biology, and cancer. We provide an overview of the role of stem cells in development, tissue homeostasis, and wound repair. We discuss the pathways and mechanisms governing stem cell plasticity and regulation of the stem cell state, and describe experimental methods for assessment of stem cells. We then review the current understanding of how environmental exposures impact stem cell function relevant to carcinogenesis and cancer prevention, with a focus on environmental and occupational exposures to chemical, physical, and biological hazards. We also highlight key areas for future research in this area, including defining whether the biological basis for cancer disparities is related to effects of complex exposure mixtures on stem cell biology.
Collapse
Affiliation(s)
- Tasha Thong
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Chanese A Forté
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA; Michigan Institute for Computational Discovery and Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Evan M Hill
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Justin A Colacino
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA; Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA; Center for Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
32
|
Ying Y, Mao Y, Yao M. NLRP3 Inflammasome Activation by MicroRNA-495 Promoter Methylation May Contribute to the Progression of Acute Lung Injury. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 18:801-814. [PMID: 31734560 PMCID: PMC6861628 DOI: 10.1016/j.omtn.2019.08.028] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 08/01/2019] [Accepted: 08/17/2019] [Indexed: 12/18/2022]
Abstract
Acute lung injury (ALI) is a pulmonary disorder that causes acute respiratory failure, thus leading to relative high mortality worldwide. However, the molecular mechanisms of ALI remain largely unknown. MicroRNA (miRNA)-dependent control of gene expression at a post-transcriptional level has been recently reported. Herein, we identify a candidate miRNA, miR-495, that affects the progression of ALI. Alveolar macrophages (NR8383) were treated with 1 μg/mL lipopolysaccharide (LPS) to establish a cell-injury model. Combined with the data from western blot, methylation-specific PCR, methylated DNA immunoprecipitation, and chromatin immunoprecipitation assays, NLRP3 inflammasome activation and methylation-dependent repression of miR-495 were found in LPS-exposed NR8383 cells. Dual-luciferase reporter gene assay and miR-495 gain-of-function experiments confirmed that NLRP3 was a target of miR-495. Next, the expression of miR-495 and NLRP3 was overexpressed or silenced to assess their effects on NLRP3 inflammasome activation, alveolar macrophage inflammation, and pyroptosis in vitro. As demonstrated, overexpressed miR-495 alleviated alveolar macrophage inflammation and pyroptosis and inhibited NLRP3 inflammasome activation by negatively regulating the NLRP3 gene. Consistently, elevated miR-495 alleviated lung injury and reduced the neutrophil infiltration and inflammation in rat models of LPS-induced ALI. Taken together, the data in our study demonstrated that methylation of the miR-495 promoter could downregulate miR-495, whose elevation could attenuate the activation of the NLRP3 inflammasome to protect against ALI, which provides novel therapeutic targets for ALI treatment.
Collapse
Affiliation(s)
- Youguo Ying
- Department of Intensive Care Unit, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201999, P.R. China
| | - Yong Mao
- Department of Intensive Care Unit, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201999, P.R. China
| | - Min Yao
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201999, P.R. China.
| |
Collapse
|
33
|
Li QC, Liang Y, Su ZB. Prophylactic treatment with MSC-derived exosomes attenuates traumatic acute lung injury in rats. Am J Physiol Lung Cell Mol Physiol 2019; 316:L1107-L1117. [PMID: 30892077 DOI: 10.1152/ajplung.00391.2018] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The mesenchymal stem cell (MSC) is a potential strategy in the pretreatment of traumatic acute lung injury (ALI), a disease that causes inflammation and oxidative stress. This study aimed to investigate whether MSC-exosomal microRNA-124-3p (miR-124-3p) affects traumatic ALI. Initially, a traumatic ALI rat model was established using the weight-drop method. Then, exosomes were obtained from MSCs of Sprague-Dawley rats, which were injected into the traumatic ALI rats. We found that miR-124-3p was abundantly-expressed in MSCs-derived exosomes and could directly target purinergic receptor P2X ligand-gated ion channel 7 (P2X7), which was overexpressed in traumatic ALI rats. After that, a loss- and gain-of-function study was performed in MSCs and traumatic ALI rats to investigate the role of miR-124-3p and P2X7 in traumatic ALI. MSC-derived exosomal miR-124-3p or silenced P2X7 was observed to increase the survival rate of traumatic ALI rats and enhance the glutathione/superoxide dismutase activity in their lung tissues. However, the wet/dry weight of lung tissues, activity of methylenedioxyamphetamine and H2O2, and levels of inflammatory factors (TNF-a, IL-6, and IL-8) were reduced. Similarly, the numbers of total cells, macrophages, neutrophils, and lymphocytes in bronchoalveolar lavage fluid were also reduced when treated with exosomal miR-124-3p or silenced P2X7. In conclusion, the results provide evidence that miR-124-3p transferred by MSC-derived exosomes inhibited P2X7 expression, thus improving oxidative stress injury and suppressing inflammatory response in traumatic ALI, highlighting a potential pretreatment for traumatic ALI.
Collapse
Affiliation(s)
- Qing-Chun Li
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
| | - Yun Liang
- Center of Physical Examination, China-Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
| | - Zhen-Bo Su
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, People’s Republic of China
| |
Collapse
|
34
|
Ye K, He D, Shao Y, Xu N, Jin C, Zhang L, Shen J. Exogenous mesenchymal stem cells affect the function of endogenous lung stem cells (club cells) in phosgene-induced lung injury. Biochem Biophys Res Commun 2019; 514:586-592. [PMID: 31064653 DOI: 10.1016/j.bbrc.2019.04.182] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 04/27/2019] [Indexed: 01/16/2023]
Abstract
Exogenous mesenchymal stem cells (MSCs) affect lung cells via cytokines as well as vesicles and activate the Notch signaling pathway thus affecting the proliferation of endogenous stem cells to repair damaged tissue. Club cells are endogenous lung stem cells whose proliferation is also closely related to the Notch signaling pathway. The club cell secretory protein (CCSP) has anti-inflammatory and anti-oxidative properties. This study aimed to investigate whether exogenous MSCs affect the function of club cells in an injured lung and whether these effects are related to the Notch signaling pathway. CCSP levels in bronchoalveolar lavage fluid (BALF) and serum were evaluated using enzyme-linked immunosorbent assay (ELISA) and the average fluorescence intensity (AFI) of CCSP in club cells was determined using flow cytometry. Immunohistochemistry and immunofluorescence were used to visualize club cells and proliferative club cells. The expression of important Notch signaling pathway components including Notch1∼4, c-myc, Hey1 and Hes1 were also assessed. LY3039478 (LY), a specific inhibitor of the Notch signaling pathway, was applied. After MSCs intervention, CCSP levels decreased, and club cell AFI increased, indicating that the secretion of club cells had weakened. The expression of Notch1, Notch2, c-myc, Hey1, Hes1 increased, accompanied by an increase in the number of proliferative club cells. Furthermore, MSCs enhanced the proliferation of club cells, while LY suppressed this phenomenon. In summary, MSCs reduced the secretion of club cells. And MSCs enhanced the proliferation of club cells partly via activating the Notch signaling pathway, which promoted lung injury repair.
Collapse
Affiliation(s)
- Kaili Ye
- Department of Intensive Care Unit, Center of Emergency and Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, China; Department of Intensive Care Unit, Medical Research Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China; Department of Intensive Care Unit, Medical Center of Radiation Injury, Jinshan Hospital, Fudan University, Shanghai, China
| | - Daikun He
- Department of Intensive Care Unit, Center of Emergency and Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, China; Department of Intensive Care Unit, Medical Research Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China; Department of Intensive Care Unit, Medical Center of Radiation Injury, Jinshan Hospital, Fudan University, Shanghai, China
| | - Yiru Shao
- Department of Intensive Care Unit, Center of Emergency and Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, China; Department of Intensive Care Unit, Medical Research Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China; Department of Intensive Care Unit, Medical Center of Radiation Injury, Jinshan Hospital, Fudan University, Shanghai, China
| | - Ning Xu
- Department of Intensive Care Unit, Center of Emergency and Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, China; Department of Intensive Care Unit, Medical Research Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China; Department of Intensive Care Unit, Medical Center of Radiation Injury, Jinshan Hospital, Fudan University, Shanghai, China
| | - Chaoyuan Jin
- Department of Intensive Care Unit, Center of Emergency and Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, China; Department of Intensive Care Unit, Medical Research Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China; Department of Intensive Care Unit, Medical Center of Radiation Injury, Jinshan Hospital, Fudan University, Shanghai, China
| | - Lin Zhang
- Department of Intensive Care Unit, Center of Emergency and Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, China; Department of Intensive Care Unit, Medical Research Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China; Department of Intensive Care Unit, Medical Center of Radiation Injury, Jinshan Hospital, Fudan University, Shanghai, China
| | - Jie Shen
- Department of Intensive Care Unit, Center of Emergency and Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, China; Department of Intensive Care Unit, Medical Research Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China; Department of Intensive Care Unit, Medical Center of Radiation Injury, Jinshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
35
|
Nie Y, Wang Z, Chai G, Xiong Y, Li B, Zhang H, Xin R, Qian X, Tang Z, Wu J, Zhao P. Dehydrocostus Lactone Suppresses LPS-induced Acute Lung Injury and Macrophage Activation through NF-κB Signaling Pathway Mediated by p38 MAPK and Akt. Molecules 2019; 24:molecules24081510. [PMID: 30999647 PMCID: PMC6514677 DOI: 10.3390/molecules24081510] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 04/15/2019] [Accepted: 04/15/2019] [Indexed: 01/10/2023] Open
Abstract
Acute lung injury (ALI) is a severe clinical disease marked by dysregulated inflammation response and has a high rate of morbidity and mortality. Macrophages, which play diverse roles in the inflammatory response, are becoming therapeutic targets in ALI. In this study we investigated the effects of dehydrocostus lactone (DHL), a natural sesquiterpene, on macrophage activation and LPS-induced ALI. The macrophage cell line RAW264.7 and primary lung macrophages were incubated with DHL (0, 3, 5, 10 and 30 μmol/L) for 0.5 h and then challenged with LPS (100 ng/mL) for up to 8 hours. C57BL/6 mice were intratracheally injected with LPS (5 mg/kg) to induce acute lung injury (ALI) and then treated with a range of DHL doses intraperitoneally (5 to 20 mg/kg). The results showed that DHL inhibited LPS-induced production of proinflammatory mediators such as iNOS, NO, and cytokines including TNF-α, IL-6, IL-1β, and IL-12 p35 by suppressing the activity of NF-κB via p38 MAPK/MK2 and Akt signaling pathway in macrophages. The in vivo results revealed that DHL significantly attenuated LPS-induced pathological injury and reduced cytokines expression in the lung. NF-κB, p38 MAPK/MK2 and Akt signaling molecules were also involved in the anti-inflammatory effect. Collectively, our findings suggested that DHL is a promising agent for alleviating LPS-induced ALI.
Collapse
Affiliation(s)
- Yunjuan Nie
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, China.
| | - Zhongxuan Wang
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, China.
| | - Gaoshang Chai
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, China.
| | - Yue Xiong
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, China.
| | - Boyu Li
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, China.
| | - Hui Zhang
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, China.
| | - Ruiting Xin
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, China.
| | - Xiaohang Qian
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, China.
| | - Zihan Tang
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, China.
| | - Jiajun Wu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, China.
| | - Peng Zhao
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi 214122, Jiangsu, China.
| |
Collapse
|
36
|
Qu L, Chen C, He W, Chen Y, Li Y, Wen Y, Zhou S, Jiang Y, Yang X, Zhang R, Shen L. Glycyrrhizic acid ameliorates LPS-induced acute lung injury by regulating autophagy through the PI3K/AKT/mTOR pathway. Am J Transl Res 2019; 11:2042-2055. [PMID: 31105816 PMCID: PMC6511780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 03/07/2019] [Indexed: 06/09/2023]
Abstract
Acute lung injury (ALI) is a major pathological issue characterized by serious inflammatory response, and a major clinically critical illness with high morbidity and mortality. Glycyrrhizic acid (GA) is a major bioactive constituent isolated from traditional Chinese herb licorice, which has been reported to have positive effects on inflammation. Nevertheless, the effects of GA on lipopolysaccharide (LPS)-treated ALI in mice have not been reported. The purpose of our study is to investigate the inhibitory effects of GA on ALI treated by LPS and to elucidate its possible mechanisms. We found that GA significantly attenuated lung injury and decreased the production of inflammatory factors TNF-α, IL-1β, and HMGB1 with LPS treatment. GA induced autophagy which was showed by enhanced number of autophagosomes through upregulating the protein levels of LC3-II/I and Beclin-1 and downregulating SQSTM1/P62. Moreover, pre-treatment of 3-Methyladenine (3-MA), an autophagy inhibitor, reversed the inhibiting effects of GA on the secretion of inflammatory factors in ALI. The PI3K/AKT/mTOR pathway was associated with GA-induced autophagy under ALI induced by LPS. In conclusion, this study indicated that GA inhibited the production of inflammatory factors in LPS-induced ALI by regulating the PI3K/AKT/mTOR pathway related autophagy, which may provide a novel therapeutic perspective of GA in ameliorating ALI.
Collapse
Affiliation(s)
- Lihua Qu
- Department of Physiology, Hunan Normal University School of MedicineChangsha 410013, China
| | - Chao Chen
- Department of Pathology and Key Laboratory of Cancer Stem Cells and Translational Medicine, Hunan Normal University School of MedicineChangsha 410013, China
| | - Wei He
- Department of Ultrasonography, The Third Xiangya Hospital of Central South UniversityChangsha 410013, China
| | - Yangye Chen
- Department of Physiology, Hunan Normal University School of MedicineChangsha 410013, China
| | - Yi Li
- Department of Physiology, Hunan Normal University School of MedicineChangsha 410013, China
| | - Yi Wen
- Department of Pharmacy and Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Hunan Normal University School of MedicineChangsha 410013, China
| | - Sichun Zhou
- Department of Pharmacy and Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Hunan Normal University School of MedicineChangsha 410013, China
| | - Yiqun Jiang
- Department of Physiology, Hunan Normal University School of MedicineChangsha 410013, China
| | - Xiaoping Yang
- Department of Pharmacy and Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Hunan Normal University School of MedicineChangsha 410013, China
| | - Ran Zhang
- Department of Immunology, Hunan Normal University School of MedicineChangsha 410013, China
| | - Li Shen
- Department of Physiology, Hunan Normal University School of MedicineChangsha 410013, China
| |
Collapse
|
37
|
Qu L, Chen C, Chen Y, Li Y, Tang F, Huang H, He W, Zhang R, Shen L. High-Mobility Group Box 1 (HMGB1) and Autophagy in Acute Lung Injury (ALI): A Review. Med Sci Monit 2019; 25:1828-1837. [PMID: 30853709 PMCID: PMC6423734 DOI: 10.12659/msm.912867] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Acute lung injury (ALI) is a life-threatening clinical syndrome in critically ill patients. The identification of novel biological markers for the early diagnosis of ALI and the development of more effective treatments are topics of current research. High mobility group box-1 protein (HMGB1) is a late inflammatory mediator associated with sepsis, malignancy, and immune disease. Levels of HMGB1 may reflect the severity of inflammation and tissue damage, indicating a potential role for HMGB1 as a prognostic biomarker in ALI, and a potential target for blocking inflammatory pathways. Several studies have shown that HMGB1 regulates autophagy. Autophagy, or type II programmed cell death, is an essential biological process that maintains cellular homeostasis. Studies have shown that HMGB1 and autophagy are involved in the pathogenesis of many lung diseases including ALI but the specific mechanisms underlying this association remain to be determined. This review aims to provide an update on the current status of the role of HMBG1 and autophagy in ALI.
Collapse
Affiliation(s)
- Lihua Qu
- Department of Physiology, Hunan Normal University Medical College, Changsha, Hunan, China (mainland)
| | - Chao Chen
- Department of Pathology and Key Laboratory of Cancer Stem Cells and Translational Medicine, Hunan Normal University Medical College, Changsha, Hunan, Christmas island
| | - YangYe Chen
- Department of Physiology, Hunan Normal University Medical College, Changsha, Hunan, China (mainland)
| | - Yi Li
- Department of Physiology, Hunan Normal University Medical College, Changsha, Hunan, China (mainland)
| | - Fang Tang
- Department of Medical Nursing, Hunan Normal University Medical College, Changsha, Hunan, China (mainland)
| | - Hao Huang
- Department of Orthopedics, The Second Affiliated Hospital of Hunan Normal University, The 163rd Central Hospital of the Peoples' Liberation Army (PLA), Changsha, Hunan, China (mainland)
| | - Wei He
- Department of Ultrasonography, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland)
| | - Ran Zhang
- Department of Immunology, Hunan Normal University Medical College, Changsha, Hunan, China (mainland)
| | - Li Shen
- Department of Physiology, Hunan Normal University Medical College, Changsha, Hunan, China (mainland)
| |
Collapse
|
38
|
Li L, Dong L, Zhang J, Gao F, Hui J, Yan J. Mesenchymal stem cells with downregulated Hippo signaling attenuate lung injury in mice with lipopolysaccharide‑induced acute respiratory distress syndrome. Int J Mol Med 2018; 43:1241-1252. [PMID: 30628652 PMCID: PMC6365074 DOI: 10.3892/ijmm.2018.4047] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 12/27/2018] [Indexed: 12/30/2022] Open
Abstract
Mesenchymal stem cell (MSC)-mediated repair of injured alveolar epithelial cells is a promising potential cure for acute respiratory distress syndrome (ARDS); however, the repairing effect of MSCs is limited by poor homing and differentiation. Our previous study revealed that the inhibition of the Hippo signaling pathway promotes the proliferation, migration and differentiation of MSCs in vitro, leading to the hypothesis that MSCs with downregulated Hippo signaling could further ameliorate lipopolysaccharide (LPS)-induced ARDS in vivo. In the current study, mouse bone marrow-derived MSCs (mMSCs) with downregulated Hippo signaling were constructed by shRNA-mediated knockdown of large tumor suppressor kinase 1 (Lats1) and were intratracheally administered to LPS-induced mouse models of ARDS. The inhibition of Hippo signaling increased the retention of mMSC in ARDS lung tissue and their differentiation toward alveolar type II epithelial cells. Furthermore, mMSCs with downregulated Hippo signaling led to a decreased lung wet weight/body weight ratio, decreased total protein and albumin concentrations in bronchoalveolar lavage fluid, decreased levels of proinflammatory factors and increased levels of anti-inflammatory factors. Finally, mMSCs with downregulated Hippo signaling improved pathological changes and decreased pulmonary fibrosis in lungs of mice with ARDS. These results suggest that the inhibition of the Hippo signaling pathway in mouse mMSCs by knockdown of Lats1 could further improve the protective effects of mMSCs against epithelial damage and the therapeutic potential of mMSCs on mice with ARDS.
Collapse
Affiliation(s)
- Lang Li
- Department of Critical Care Medicine, Wuxi People's Hospital, Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| | - Liang Dong
- Department of Critical Care Medicine, Wuxi People's Hospital, Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| | - Jiangqian Zhang
- Department of Critical Care Medicine, Wuxi People's Hospital, Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| | - Fei Gao
- Department of Critical Care Medicine, Wuxi People's Hospital, Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| | - Jiaojie Hui
- Department of Critical Care Medicine, Wuxi People's Hospital, Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| | - Jie Yan
- Department of Critical Care Medicine, Wuxi People's Hospital, Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| |
Collapse
|
39
|
Conese M, Beccia E, Carbone A, Castellani S, Di Gioia S, Corti F, Angiolillo A, Colombo C. The role of stem cells in cystic fibrosis disease modeling and drug discovery. Expert Opin Orphan Drugs 2018. [DOI: 10.1080/21678707.2018.1549480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Massimo Conese
- Laboratory of Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Elisa Beccia
- Laboratory of Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso, Italy
| | - Annalucia Carbone
- Division of Internal Medicine and Chronobiology Unit, IRCCS “Casa Sollievo della Sofferenza”, San Giovanni Rotondo (FG), Italy
| | - Stefano Castellani
- Laboratory of Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Sante Di Gioia
- Laboratory of Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Fabiola Corti
- Department of Pathophysiology and Transplantation, Cystic Fibrosis Center, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Antonella Angiolillo
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso, Italy
| | - Carla Colombo
- Department of Pathophysiology and Transplantation, Cystic Fibrosis Center, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| |
Collapse
|
40
|
Sun Z, Li F, Zhou X, Chung KF, Wang W, Wang J. Stem cell therapies for chronic obstructive pulmonary disease: current status of pre-clinical studies and clinical trials. J Thorac Dis 2018; 10:1084-1098. [PMID: 29607186 DOI: 10.21037/jtd.2018.01.46] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a respiratory disease that has a major impact worldwide. The currently-available drugs mainly focus on relieving the symptoms of COPD patients. Novel regenerative therapeutic approaches have been investigated with the aim of repairing or replacing the injured functional structures of the respiratory system. We summarized the progress made by regenerative therapies for COPD by analyzing results from both pre-clinical studies and completed clinical trials. These approaches include the application of exogenous stem cells or small molecules to stimulate the regeneration by endogenous lung stem/progenitor cells. Exogenous mesenchymal stem cells (MSCs) have been reported to repair the structure and improve the function of the injured respiratory system in COPD models. However, the studies that used MSCs in patients with moderate-to-severe COPD patients did not lead to clear respiratory functional improvements. Exogenous human lung stem cells applied to cryo-injured (CI) lungs of mice have been shown to organize into human-like pulmonary structures, indicating a new property of stem cells that is potentially capable of curing COPD patients. Small molecules like retinoic acid has been shown to lead to regeneration and repair of the damaged lung structures in COPD mouse models probably by activation of endogenous lung stem/progenitor cells. However, retinoic acid or agonists of retinoic acid receptor administered to moderate or severe COPD patients did not improve the density and function of the damaged lung. These novel regenerative approaches have failed in preliminary clinical trials, possibly due to the advanced severity of the disease. Further work should be done to develop the current regenerative approaches for curing patients at different stages of COPD. We suggest that some modifications of the approach in the clinical studies may lead to more successful outcomes of regenerative therapy for COPD.
Collapse
Affiliation(s)
- Zhongwei Sun
- Cellular Biomedicine Group, Shanghai 200233, China.,Cellular Biomedicine Group, Cupertino, CA, USA
| | - Feng Li
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai 200080, China
| | - Xin Zhou
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai 200080, China
| | - Kian Fan Chung
- Airway Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Wen Wang
- Cellular Biomedicine Group, Shanghai 200233, China.,Cellular Biomedicine Group, Cupertino, CA, USA
| | - Jialun Wang
- Cellular Biomedicine Group, Shanghai 200233, China.,Cellular Biomedicine Group, Cupertino, CA, USA
| |
Collapse
|
41
|
Guo Q, Wang J. Effect of combination of vitamin E and umbilical cord-derived mesenchymal stem cells on inflammation in mice with acute kidney injury. Immunopharmacol Immunotoxicol 2018; 40:168-172. [PMID: 29355065 DOI: 10.1080/08923973.2018.1424898] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND The objective of this study is to investigate the effect of combination of umbilical cord-derived mesenchymal stem cell (UC-MSC) and vitamin E (VitE) on inflammation in mice with acute kidney injury (AKI). METHODS UC-MSCs were isolated from pregnant wistar mice and cultured. A total of 90 female wistar mice were randomly divided into control group, AKI group, AKI + VitE group, AKI + UC-MSC group, and AKI + VitE + UC-MSC group (18 mice in each group) which were given no treatment, normal saline, VitE, UC-MSC, and VitE + UC-MSC, respectively. The renal pedicles on both sides were clipped for 50 min with micro-artery clips to induce AKI. Six mice were sacrificed at days 1, 3, and 7, while blood and kidney tissues were collected to detect levels of blood urea nitrogen (BUN) and creatinine (Scr). Kidney tissues were stained by HE staining to observe pathological changes; levels of interleukin-lβ, TNF-α, interleukin-10, and β-FGF were measured by ELISA. RESULTS Compared with the control group, AKI mice showed higher levels of serum BUN and Scr, tubular swelling and necrosis suggesting that AKI model was successfully established. Mice in AKI + VitE group, AKI + UC-MSC group, and AKI + VitE + UC-MSC presented better renal function than mice of AKI group. Mice from AKI + VitE + UC-MSC group showed the best renal function with the least renal tubular injury (p < .05). ELISA detection revealed that pro-inflammatory cytokines were significantly increased and anti-inflammatory cytokine levels were significantly decreased in all time points (p < .05). VitE, UC-MSC, and VitE + UC-MSC resulted in the increase of anti-inflammatory cytokine levels and reduction of pro-inflammatory cytokine levels and the combination of VitE and UC-MSC performed favorable effect in the suppression of inflammation in AKI mice (p < .05). CONCLUSIONS Combination of UC-MSC and VitE significantly inhibited inflammatory reaction in kidney through the regulation of inflammatory cytokines in the microenvironment of kidney with AKI. Combination of UC-MSC and VitE presented therapeutic effect on AKI than the single use of UC-MSC or VitE.
Collapse
Affiliation(s)
- Qiongqiong Guo
- a Department of Hemopurification Center , The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology , Luoyang , China
| | - Junxia Wang
- a Department of Hemopurification Center , The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology , Luoyang , China
| |
Collapse
|
42
|
Gap Junctions Are Involved in the Rescue of CFTR-Dependent Chloride Efflux by Amniotic Mesenchymal Stem Cells in Coculture with Cystic Fibrosis CFBE41o- Cells. Stem Cells Int 2018. [PMID: 29531530 PMCID: PMC5821953 DOI: 10.1155/2018/1203717] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
We previously found that human amniotic mesenchymal stem cells (hAMSCs) in coculture with CF immortalised airway epithelial cells (CFBE41o- line, CFBE) on Transwell® filters acquired an epithelial phenotype and led to the expression of a mature and functional CFTR protein. In order to explore the role of gap junction- (GJ-) mediated intercellular communication (GJIC) in this rescue, cocultures (hAMSC : CFBE, 1 : 5 ratio) were studied for the formation of GJIC, before and after silencing connexin 43 (Cx43), a major component of GJs. Functional GJs in cocultures were inhibited when the expression of the Cx43 protein was downregulated. Transfection of cocultures with siRNA against Cx43 resulted in the absence of specific CFTR signal on the apical membrane and reduction in the mature form of CFTR (band C), and in parallel, the CFTR-dependent chloride channel activity was significantly decreased. Cx43 downregulation determined also a decrease in transepithelial resistance and an increase in paracellular permeability as compared with control cocultures, implying that GJIC may regulate CFTR expression and function that in turn modulate airway epithelium tightness. These results indicate that GJIC is involved in the correction of CFTR chloride channel activity upon the acquisition of an epithelial phenotype by hAMSCs in coculture with CF cells.
Collapse
|
43
|
Conese M, Beccia E, Castellani S, Di Gioia S, Colombo C, Angiolillo A, Carbone A. The long and winding road: stem cells for cystic fibrosis. Expert Opin Biol Ther 2017; 18:281-292. [PMID: 29216777 DOI: 10.1080/14712598.2018.1413087] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Cystic fibrosis (CF) is a genetic syndrome with a high mortality rate due to severe lung disease. Despite having several drugs targeting specific mutated CFTR proteins already in clinical trials, new therapies, based on stem cells, are also emerging to treat those patients. AREAS COVERED The authors review the main sources of stem cells, including embryonic stem cells (ESCs), induced-pluripotent stem cells (iPSCs), gestational stem cells, and adult stem cells, such as mesenchymal stem cells (MSCs) in the context of CF. Furthermore, they describe the main animal and human models of lung physiology and pathology, involved in the optimization of these stem cell-applied therapies in CF. EXPERT OPINION ESCs and iPSCs are emerging sources for disease modeling and drug discovery purposes. The allogeneic transplant of healthy MSCs, that acts independently to specific mutations, is under intense scrutiny due to their secretory, immunomodulatory, anti-inflammatory and anti-bacterial properties. The main challenge for future developments will be to get exogenous stem cells into the appropriate lung location, where they can regenerate endogenous stem cells and act as inflammatory modulators. The clinical application of stem cells for the treatment of CF certainly warrants further insight into pre-clinical models, including large animals, organoids, decellularized organs and lung bioengineering.
Collapse
Affiliation(s)
- Massimo Conese
- a Laboratory of Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences , University of Foggia , Foggia , Italy
| | - Elisa Beccia
- a Laboratory of Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences , University of Foggia , Foggia , Italy.,b Department of Medicine and Health Sciences 'V. Tiberio' , University of Molise , Campobasso , Italy
| | - Stefano Castellani
- a Laboratory of Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences , University of Foggia , Foggia , Italy
| | - Sante Di Gioia
- a Laboratory of Experimental and Regenerative Medicine, Department of Medical and Surgical Sciences , University of Foggia , Foggia , Italy
| | - Carla Colombo
- c Cystic Fibrosis Center, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Department of Pathophysiology and Transplantation , University of Milan , Milan , Italy
| | - Antonella Angiolillo
- b Department of Medicine and Health Sciences 'V. Tiberio' , University of Molise , Campobasso , Italy
| | - Annalucia Carbone
- d Division of Internal Medicine and Chronobiology Unit , IRCCS 'Casa Sollievo della Sofferenza' , San Giovanni Rotondo (FG) , Italy
| |
Collapse
|
44
|
Wu DQ, Wu HB, Zhang M, Wang JA. Effects of Zinc Finger Protein A20 on Lipopolysaccharide (LPS)-Induced Pulmonary Inflammation/Anti-Inflammatory Mediators in an Acute Lung Injury/Acute Respiratory Distress Syndrome Rat Model. Med Sci Monit 2017; 23:3536-3545. [PMID: 28724884 PMCID: PMC5533196 DOI: 10.12659/msm.901700] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Background The aim of this study was to investigate the effects of zinc finger protein A20 on lipopolysaccharide (LPS)-induced pulmonary inflammation/anti-inflammatory mediators in an acute lung injury/acute respiratory distress syndrome (ALI/ARDS) rat model. Material/Methods Forty-eight ALI/ARDS rats were selected and assigned into normal saline (NS) (injected with NS), LPS (injected with LPS), LPS-C1 (injected with pEGFP-C1, NS and LPS), and A20 groups (injected with pEGFP-C1-A20, NS, and LPS). The wet/dry (W/D) ratio of rat lung tissues and total protein concentration and the number of neutrophils in bronchoalveolar lavage fluid (BALF) were detected. Enzyme-linked immunosorbent assay (ELISA) and qRT-PCR were applied to detect the protein and mRNA expressions of A20, IL-10, and TNF-α, respectively. Western blotting was employed to detect the protein expressions of A20, nuclear factor-kappa B (NF-κB) p65 and NF-κB p-P65 in rat lung tissues. Results Compared with the NS group, the W/D ratio of rat lung tissues and total protein concentration and the number of neutrophils in BALF in the other 3 groups increased significantly. The protein and mRNA expressions of A20, IL-10, and TNF-α were significantly higher in the LPS group than in the NS group. The protein and mRNA expressions of A20 and IL-10 were significantly up-regulated and the expression of TNF-α, NF-κB p65, and NF-κB p-P65 was significantly down-regulated in rats injected with A20 compared to those in the LPS group. Conclusions The study provided evidence that zinc finger protein A20 can alleviate pulmonary inflammation by inhibiting TNF-α, NF-κB p65, and NF-κB p-P65 expressions and promoting IL-10 expression.
Collapse
Affiliation(s)
- Ding-Qian Wu
- Department of Emergency, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China (mainland)
| | - Hong-Bo Wu
- Department of Emergency, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China (mainland)
| | - Mao Zhang
- Department of Emergency, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China (mainland)
| | - Jian-An Wang
- Department of Internal Medicine-Cardiovascular, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China (mainland)
| |
Collapse
|
45
|
The Use of GMP-Produced Bone Marrow-Derived Stem Cells in Combination with Extracorporeal Membrane Oxygenation in ARDS: An Animal Model. ASAIO J 2017; 63:324-332. [DOI: 10.1097/mat.0000000000000566] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
46
|
Dynamic Tracking Human Mesenchymal Stem Cells Tropism following Smoke Inhalation Injury in NOD/SCID Mice. Stem Cells Int 2016; 2016:1691856. [PMID: 27725837 PMCID: PMC5048056 DOI: 10.1155/2016/1691856] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 06/22/2016] [Accepted: 06/28/2016] [Indexed: 12/12/2022] Open
Abstract
Multiple preclinical evidences have supported the potential value of mesenchymal stem cells (MSCs) for treatment of acute lung injury (ALI). However, few studies focus on the dynamic tropism of MSCs in animals with acute lung injury. In this study, we track systemically transplanted human bone marrow-derived mesenchymal stem cells (hBMSCs) in NOD/SCID mice with smoke inhalation injury (SII) through bioluminescence imaging (BLI). The results showed that hBMSCs systemically delivered into healthy NOD/SCID mouse initially reside in the lungs and then partially translocate to the abdomen after 24 h. Compared with the uninjured control group treated with hBMSCs, higher numbers of hBMSCs were found in the lungs of the SII NOD/SCID mice. In both the uninjured and SII mice, the BLI signals in the lungs steadily decreased over time and disappeared by 5 days after treatment. hBMSCs significantly attenuated lung injury, elevated the levels of KGF, decreased the levels of TNF-α in BALF, and inhibited inflammatory cell infiltration in the mice with SII. In conclusion, our findings demonstrated that more systemically infused hBMSCs localized to the lungs in mice with SII. hBMSC xenografts repaired smoke inhalation-induced lung injury in mice. This repair was maybe due to the effect of anti-inflammatory and secreting KGF of hMSCs but not associated with the differentiation of the hBMSCs into alveolar epithelial cells.
Collapse
|
47
|
Hegab AE, Betsuyaku T. Lung Stem Cells and Their Use for Patient Care: Are We There Yet? ACTA ACUST UNITED AC 2016. [DOI: 10.1007/978-3-319-33270-3_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
48
|
Xu Y, Xiang J, Zhao H, Liang H, Huang J, Li Y, Pan J, Zhou H, Zhang X, Wang JH, Liu Z, Wang J. Human amniotic fluid stem cells labeled with up-conversion nanoparticles for imaging-monitored repairing of acute lung injury. Biomaterials 2016; 100:91-100. [PMID: 27244692 DOI: 10.1016/j.biomaterials.2016.05.034] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 04/26/2016] [Accepted: 05/17/2016] [Indexed: 01/11/2023]
Abstract
Human amniotic fluid stem (hAFS) cells have generated a great deal of excitement in cell-based therapies and regenerative medicine. Here, we examined the effect of hAFS cells labeled with dual-polymer-coated UCNP-PEG-PEI nanoparticles in a murine model of acute lung injury (ALI). We observed hAFS cells migration to the lung using highly sensitive in vivo upconversion luminescence (UCL) imaging. We demonstrated that hAFS cells remained viable and retained their ability to differentiate even after UCNP-PEG-PEI labeling. More importantly, hAFS cells displayed remarkable positive effects on ALI-damaged lung tissue repair compared with mouse bone marrow mesenchymal stem cells (mBMSCs), which include recovery of the integrity of alveolar-capillary membrane, attenuation of transepithelial leukocyte and neutrophil migration, and down-regulation of proinflammatory cytokine and chemokine expression. Our work highlights a promising role for imaging-guided hAFS cell-based therapy in ALI.
Collapse
Affiliation(s)
- Yunyun Xu
- Institute for Pediatric Research, Affiliated Children's Hospital, Soochow University, Suzhou, Jiangsu 215025, China
| | - Jian Xiang
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu 215123, China
| | - He Zhao
- Institute for Pediatric Research, Affiliated Children's Hospital, Soochow University, Suzhou, Jiangsu 215025, China
| | - Hansi Liang
- Stem Cell and Biomedical Material Key Laboratory of Jiangsu Province, State Key Laboratory Incubation Base, Soochow University, Suzhou, Jiangsu 215007, China
| | - Jie Huang
- Institute for Pediatric Research, Affiliated Children's Hospital, Soochow University, Suzhou, Jiangsu 215025, China
| | - Yan Li
- Institute for Pediatric Research, Affiliated Children's Hospital, Soochow University, Suzhou, Jiangsu 215025, China
| | - Jian Pan
- Institute for Pediatric Research, Affiliated Children's Hospital, Soochow University, Suzhou, Jiangsu 215025, China
| | - Huiting Zhou
- Institute for Pediatric Research, Affiliated Children's Hospital, Soochow University, Suzhou, Jiangsu 215025, China
| | - Xueguang Zhang
- Stem Cell and Biomedical Material Key Laboratory of Jiangsu Province, State Key Laboratory Incubation Base, Soochow University, Suzhou, Jiangsu 215007, China
| | - Jiang Huai Wang
- Department of Academic Surgery, University College Cork, Cork University Hospital, Cork, Ireland
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Jian Wang
- Institute for Pediatric Research, Affiliated Children's Hospital, Soochow University, Suzhou, Jiangsu 215025, China.
| |
Collapse
|
49
|
Monsel A, Zhu YG, Gudapati V, Lim H, Lee JW. Mesenchymal stem cell derived secretome and extracellular vesicles for acute lung injury and other inflammatory lung diseases. Expert Opin Biol Ther 2016; 16:859-71. [PMID: 27011289 DOI: 10.1517/14712598.2016.1170804] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Acute respiratory distress syndrome is a major cause of respiratory failure in critically ill patients. Despite extensive research into its pathophysiology, mortality remains high. No effective pharmacotherapy exists. Based largely on numerous preclinical studies, administration of mesenchymal stem or stromal cell (MSC) as a therapeutic for acute lung injury holds great promise, and clinical trials are currently underway. However, concern for the use of stem cells, specifically the risk of iatrogenic tumor formation, remains unresolved. Accumulating evidence now suggest that novel cell-free therapies including MSC-derived conditioned medium and extracellular vesicles released from MSCs might constitute compelling alternatives. AREAS COVERED The current review summarizes the preclinical studies testing MSC conditioned medium and/or MSC extracellular vesicles as treatment for acute lung injury and other inflammatory lung diseases. EXPERT OPINION While certain logistical obstacles limit the clinical applications of MSC conditioned medium such as the volume required for treatment, the therapeutic application of MSC extracellular vesicles remains promising, primarily due to ability of extracellular vesicles to maintain the functional phenotype of the parent cell. However, utilization of MSC extracellular vesicles will require large-scale production and standardization concerning identification, characterization and quantification.
Collapse
Affiliation(s)
- Antoine Monsel
- a Multidisciplinary Intensive Care Unit, Department of Anesthesiology and Critical Care , La Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris, University Pierre and Marie Curie (UPMC) Univ Paris 06 , Paris , France
| | - Ying-Gang Zhu
- b Department of Pulmonary Disease , Huadong Hospital, Fudan University , Shanghai , China
| | - Varun Gudapati
- c Department of Anesthesiology , University of California San Francisco , San Francisco , CA , USA
| | - Hyungsun Lim
- c Department of Anesthesiology , University of California San Francisco , San Francisco , CA , USA
| | - Jae W Lee
- c Department of Anesthesiology , University of California San Francisco , San Francisco , CA , USA
| |
Collapse
|
50
|
Bajaj P, Harris JF, Huang JH, Nath P, Iyer R. Advances and Challenges in Recapitulating Human Pulmonary Systems: At the Cusp of Biology and Materials. ACS Biomater Sci Eng 2016; 2:473-488. [PMID: 33465851 DOI: 10.1021/acsbiomaterials.5b00480] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The aim of this review is to provide an overview of physiologically relevant microengineered lung-on-a-chip (LoC) platforms for a variety of different biomedical applications with emphasis on drug screening. First, a brief outline of lung anatomy and physiology is presented followed by discussion of the lung parenchyma and its extracellular matrix. Next, we point out the technical challenges in recapitulating the complexity of lung in conventional static two-dimensional microenvironments and the need for alternate lung platforms. The importance of scaling laws is also emphasized in designing these in vitro microengineered lung platforms. The review then discusses current LoC platforms that have been used for testing the efficacy of drugs or as model systems for investigating disorders of the lung parenchyma. Finally, the design parameters in developing an ideal physiologically relevant LoC platform are presented. As this emerging field of organ-on-a-chip can serve an alternative platform for animal testing of drugs or modeling human diseases in vitro, it has significant potential to impact the future of pharmaceutical research.
Collapse
Affiliation(s)
- Piyush Bajaj
- Information Systems and Modeling, §Bioscience Division, and ⊥Physics Division, Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
| | - Jennifer F Harris
- Information Systems and Modeling, Bioscience Division, and ⊥Physics Division, Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
| | - Jen-Huang Huang
- Information Systems and Modeling, Bioscience Division, and Physics Division, Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
| | - Pulak Nath
- Information Systems and Modeling, Bioscience Division, and Physics Division, Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
| | - Rashi Iyer
- Information Systems and Modeling, Bioscience Division, and Physics Division, Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
| |
Collapse
|