1
|
Karpurapu M, Nie Y, Chung S, Yan J, Dougherty P, Pannu S, Wisler J, Harkless R, Parinandi N, Berdyshev E, Pei D, Christman JW. The calcineurin-NFATc pathway modulates the lipid mediators in BAL fluid extracellular vesicles, thereby regulating microvascular endothelial cell barrier function. Front Physiol 2024; 15:1378565. [PMID: 38812883 PMCID: PMC11133699 DOI: 10.3389/fphys.2024.1378565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/16/2024] [Indexed: 05/31/2024] Open
Abstract
Extracellular vesicles mediate intercellular communication by transporting biologically active macromolecules. Our prior studies have demonstrated that the nuclear factor of activated T cell cytoplasmic member 3 (NFATc3) is activated in mouse pulmonary macrophages in response to lipopolysaccharide (LPS). Inhibition of NFATc3 activation by a novel cell-permeable calcineurin peptide inhibitor CNI103 mitigated the development of acute lung injury (ALI) in LPS-treated mice. Although pro-inflammatory lipid mediators are known contributors to lung inflammation and injury, it remains unclear whether the calcineurin-NFATc pathway regulates extracellular vesicle (EV) lipid content and if this content contributes to ALI pathogenesis. In this study, EVs from mouse bronchoalveolar lavage fluid (BALF) were analyzed for their lipid mediators by liquid chromatography in conjunction with mass spectrometry (LC-MS/MS). Our data demonstrate that EVs from LPS-treated mice contained significantly higher levels of arachidonic acid (AA) metabolites, which were found in low levels by prior treatment with CNI103. The catalytic activity of lung tissue cytoplasmic phospholipase A2 (cPLA2) increased during ALI, correlating with an increased amount of arachidonic acid (AA) in the EVs. Furthermore, ALI is associated with increased expression of cPLA2, cyclooxygenase 2 (COX2), and lipoxygenases (5-LOX, 12-LOX, and 15-LOX) in lung tissue, and pretreatment with CNI103 inhibited the catalytic activity of cPLA2 and the expression of cPLA2, COX, and LOX transcripts. Furthermore, co-culture of mouse pulmonary microvascular endothelial cell (PMVEC) monolayer and NFAT-luciferase reporter macrophages with BALF EVs from LPS-treated mice increased the pulmonary microvascular endothelial cell (PMVEC) monolayer barrier permeability and luciferase activity in macrophages. However, EVs from CNI103-treated mice had no negative impact on PMVEC monolayer barrier integrity. In summary, BALF EVs from LPS-treated mice carry biologically active NFATc-dependent, AA-derived lipids that play a role in regulating PMVEC monolayer barrier function.
Collapse
Affiliation(s)
- Manjula Karpurapu
- Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, United States
| | - Yunjuan Nie
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Sangwoon Chung
- Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, United States
| | - Jiasheng Yan
- Department of Pharmacology, Ohio State University, Columbus, OH, United States
| | - Patrick Dougherty
- Department of Chemistry and Biochemistry, Ohio State University, Columbus, OH, United States
| | - Sonal Pannu
- Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, United States
| | - Jon Wisler
- Department of Surgery, Ohio State Wexner Medical Center, Columbus, OH, United States
| | - Ryan Harkless
- Department of Surgery, Ohio State Wexner Medical Center, Columbus, OH, United States
| | - Narasimham Parinandi
- Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, United States
| | - Evgeny Berdyshev
- Division of Pulmonary Critical Care and Sleep Medicine, National Jewish Health, Denver, CO, United States
| | - Dehua Pei
- Department of Chemistry and Biochemistry, Ohio State University, Columbus, OH, United States
| | - John W. Christman
- Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, United States
| |
Collapse
|
2
|
Zhong XQ, Hao TF, Zhu QJ, Zheng J, Zheng MF, Li XH, Luo LH, Xia CS, Fan YW, Gu J, Liu T, Chen DJ. Umbilical cord blood exosomes from very preterm infants with bronchopulmonary dysplasia aggravate lung injury in mice. Sci Rep 2023; 13:8648. [PMID: 37244977 DOI: 10.1038/s41598-023-35620-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/21/2023] [Indexed: 05/29/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) is characterized by abnormal development of the blood vessels and alveoli in lungs, which largely occurs in premature infants. Exosomes (EXO) from very preterm infants (VPI) with BPD (BPD-EXO) impair angiogenic activities of human umbilical vein endothelial cells (HUVECs) via EXO-miRNAs cargo. This study aimed to determine whether and how BPD-EXO affect the development of BPD in a mouse model. We showed that treating BPD mice with BPD-EXO chronically and irreversibly aggravated lung injury. BPD-EXO up-regulated 139 and down-regulated 735 genes in the mouse lung tissue. These differentially expressed genes were enriched to the MAPK pathway (e.g., Fgf9 and Cacna2d3), which is critical to angiogenesis and vascular remodeling. BPD-EXO suppressed expression of Fgf9 and Cacna2d3 in HUVECs and inhibited migration, tube formation, and increased cell apoptosis in HUVECs. These data demonstrate that BPD-EXO aggravate lung injury in BPD mice and impair lung angiogenesis, plausibly leading to adverse outcomes of VPI with BPD. These data also suggest that BPD-EXO could serve as promising targets for predicting and treating BPD.
Collapse
Affiliation(s)
- Xin-Qi Zhong
- Department of Neonatology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Liwan District, Guangzhou, 510150, China.
- Key Laboratory for Major Obstetric Disease of Guangdong Province, Guangzhou, China.
| | - Tao-Fang Hao
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Qi-Jiong Zhu
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, 510632, China
- China Greater Bay Area Research Center of Environmental Health, School of Medicine, Jinan University, Guangzhou, China
| | - Jing Zheng
- Department of Obstetrics and Gynecology, University of WI-Madison, Madison, WI, USA
| | - Mao-Fei Zheng
- Department of Neonatology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Liwan District, Guangzhou, 510150, China
| | - Xiu-Hong Li
- Department of Maternal and Child Health, School of Public Health, Sun Yat-Sen University, Guangzhou, China
| | - Li-Hua Luo
- Department of Neonatology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Liwan District, Guangzhou, 510150, China
| | - Chang-Shun Xia
- Department of Neonatology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Liwan District, Guangzhou, 510150, China
| | - Yu-Wei Fan
- Department of Neonatology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Liwan District, Guangzhou, 510150, China
| | - Jian Gu
- Department of Neonatology, The Third Affiliated Hospital of Guangzhou Medical University, 63 Duobao Road, Liwan District, Guangzhou, 510150, China
| | - Tao Liu
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, 510632, China.
- China Greater Bay Area Research Center of Environmental Health, School of Medicine, Jinan University, Guangzhou, China.
| | - Dun-Jin Chen
- Key Laboratory for Major Obstetric Disease of Guangdong Province, Guangzhou, China.
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, Guangdong, China.
| |
Collapse
|
3
|
Wang J, Zhang A, Huang F, Xu J, Zhao M. MSC-EXO and tempol ameliorate bronchopulmonary dysplasia in newborn rats by activating HIF-1α. Pediatr Pulmonol 2023; 58:1367-1379. [PMID: 36650825 DOI: 10.1002/ppul.26317] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 12/25/2022] [Accepted: 01/15/2023] [Indexed: 01/19/2023]
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) is a major complication of premature infants and an important cause of morbidity and mortality. This study investigates the effect of the combination of mesenchymal stem cells-derived exosomes (MSC-EXO) and tempol on BPD and analyzes its mechanism. METHODS MSC-EXO was extracted by centrifugation and identified by transmission electron microscopy (TEM), nanoparticle tracking analysis, and western blot analysis (WB). Tidal volume (TV), minute ventilation (MV), peak inspiratory flow (PIF), and dynamic pulmonary compliance (Cdyn) of rats were measured by BuxCo pulmonary function experimental platform. Hematoxylin-eosin staining was performed to observe the lung morphology and radical alveolar count (RAC) and mean linear intercept (MLI) were assessed. Immunofluorescence (IF) was conducted to detect the expression of CD31 and α-SMA in pulmonary blood vessels. The kits were used to calculate malondialdehyde (MDA), superoxide dismutase (SOD), and total antioxidant capacity (TAOC) concentration in lung tissue. Enzyme linked immunosorbent assay was applied to detect the levels of IL-1β, IL-17, IL-6, and IFN-γ in bronchoalveolar lavage fluid. In addition, the expressions of HIF-1α, vascular endothelial growth factor (VEGF), p-PI3K, and p-AKT were analyzed by WB and IF. RESULTS We successfully extracted and identified MSC-EXO. In BPD rats, TV, MV, PIF, and Cdyn decreased, alveoli were simplified, and the number of interalveoli small vessels, blood vessel density decreased. Moreover, RAC, CD31, TAOC, and SOD decreased, and MLI, α-SMA, MDA, IL-1β, IL-17, IL-6, and IFN-γ increased, which was reversed by the combination of MSC-EXO and tempol treatment after combined treatment. In addition, the expression levels of HIF-1α, VEGF, p-PI3K, and p-AKT were increased after combined treatment. CONCLUSIONS Combined treatment could improve lung tissue injury, promote pulmonary vascular remodeling, restore lung function, and inhibit oxidative stress in BPD rats. These effects were achieved through activation of HIF-1α.
Collapse
Affiliation(s)
- Juanmei Wang
- Department of Pediatrics, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, China.,Hunan Provincial Key Laboratory of Pediatric Respirology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Aimin Zhang
- Department of Pediatrics, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Furong Huang
- Department of Pediatrics, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Jun Xu
- Department of Pediatrics, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Menghua Zhao
- Department of Pediatrics, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, China
| |
Collapse
|
4
|
Xi Y, Ju R, Wang Y. Mesenchymal Stem Cell-Derived Extracellular Vesicles for the Treatment of Bronchopulmonary Dysplasia. Front Pediatr 2022; 10:852034. [PMID: 35444971 PMCID: PMC9013803 DOI: 10.3389/fped.2022.852034] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/25/2022] [Indexed: 11/13/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the most common chronic respiratory disease in premature infants. However, there is a lack of effective treatment. Mesenchymal stromal cells derived extracellular vesicles (MSC-EVs), as nano- and micron-sized heterogeneous vesicles secreted by MSCs, are the main medium for information exchange between MSCs and injured tissue and organ, playing an important role in repairing tissue and organ injury. EVs include exosomes, microvesicles and so on. They are rich with various proteins, nucleic acids, and lipids. Now, EVs are considered as a new way of cell-to-cell communication. EVs mainly induce regeneration and therapeutic effects in different tissues and organs through the biomolecules they carry. The surface membrane protein or loaded protein and nucleic acid molecules carried by EVs, can activate the signal transduction of target cells and regulate the biological behavior of target cells after binding and cell internalization. MSC-EVs can promote the development of pulmonary vessels and alveoli and reduce pulmonary hypertension (PH) and inflammation and play an important role in the repair of lung injury in BPD. The regeneration potential of MSC-EVs is mainly due to the regulation of cell proliferation, survival, migration, differentiation, angiogenesis, immunoregulation, anti-inflammatory, mitochondrial activity and oxidative stress. As a new type of cell-free therapy, MSC-EVs have non-immunogenic, and are small in size and go deep into most tissues. What's more, it has good biological stability and can be modified and loaded with drugs of interest. Obviously, MSC-EVs have a good application prospect in the treatment of lung injury and BPD. However, there are still many challenges to make MSC-EVs really enter clinical application.
Collapse
Affiliation(s)
- Yufeng Xi
- Department of Neonatology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Rong Ju
- Department of Neonatology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yujia Wang
- Department of Neonatology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.,Department of Dermatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
5
|
Zhong XQ, Yan Q, Chen ZG, Jia CH, Li XH, Liang ZY, Gu J, Wei HL, Lian CY, Zheng J, Cui QL. Umbilical Cord Blood-Derived Exosomes From Very Preterm Infants With Bronchopulmonary Dysplasia Impaired Endothelial Angiogenesis: Roles of Exosomal MicroRNAs. Front Cell Dev Biol 2021; 9:637248. [PMID: 33842462 PMCID: PMC8027316 DOI: 10.3389/fcell.2021.637248] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 02/22/2021] [Indexed: 12/13/2022] Open
Abstract
Premature infants have a high risk of bronchopulmonary dysplasia (BPD), which is characterized by abnormal development of alveoli and pulmonary vessels. Exosomes and exosomal miRNAs (EXO-miRNAs) from bronchoalveolar lavage fluid are involved in the development of BPD and might serve as predictive biomarkers for BPD. However, the roles of exosomes and EXO-miRNAs from umbilical cord blood of BPD infants in regulating angiogenesis are yet to be elucidated. In this study, we showed that umbilical cord blood-derived exosomes from BPD infants impaired angiogenesis in vitro. Next-generation sequencing of EXO-miRNAs from preterm infants without (NBPD group) or with BPD (BPD group) uncovered a total of 418 differentially expressed (DE) EXO-miRNAs. These DE EXO-miRNAs were primarily enriched in cellular function-associated pathways including the PI3K/Akt and angiogenesis-related signaling pathways. Among those EXO-miRNAs which are associated with PI3K/Akt and angiogenesis-related signaling pathways, BPD reduced the expression of hsa-miR-103a-3p and hsa-miR-185-5p exhibiting the most significant reduction (14.3% and 23.1% of NBPD group, respectively); BPD increased hsa-miR-200a-3p expression by 2.64 folds of the NBPD group. Furthermore, overexpression of hsa-miR-103a-3p and hsa-miR-185-5p in normal human umbilical vein endothelial cells (HUVECs) significantly enhanced endothelial cell proliferation, tube formation, and cell migration, whereas overexpressing hsa-miR-200a-3p inhibited these cellular responses. This study demonstrates that exosomes derived from umbilical cord blood of BPD infants impair angiogenesis, possibly via DE EXO-miRNAs, which might contribute to the development of BPD.
Collapse
Affiliation(s)
- Xin-Qi Zhong
- Department of Neonatology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Key Laboratory for Major Obstetric Diseases of Guangdong Province, Guangzhou, China
| | - Qin Yan
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhuang-Gui Chen
- Department of Pediatrics and Department of Allergy, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chun-Hong Jia
- Department of Neonatology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiu-Hong Li
- Department of Maternal and Child Health, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Zi-Yan Liang
- Department of Neonatology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jian Gu
- Department of Neonatology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hui-Ling Wei
- Department of Neonatology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chang-Yu Lian
- Department of Neonatology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jing Zheng
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
| | - Qi-Liang Cui
- Department of Neonatology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Key Laboratory for Major Obstetric Diseases of Guangdong Province, Guangzhou, China
| |
Collapse
|
6
|
Qian X, An N, Ren Y, Yang C, Zhang X, Li L. Immunosuppressive Effects of Mesenchymal Stem Cells-derived Exosomes. Stem Cell Rev Rep 2020; 17:411-427. [PMID: 32935222 DOI: 10.1007/s12015-020-10040-7] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Mesenchymal stem cells (MSCs) have become important seed cells in therapy because of their immunosuppressive function and anti-inflammatory effects. MSCs exert immunosuppressive effects through direct contact or paracrine action. The paracrine functions of MSCs are at least partially mediated by exosomes, which are membrane vesicles, carrying abundant proteins, nucleic acids and other active molecules. MSC-exos have heterogeneity. The exosomes from different donors, tissues generations of MSCs carry different bioactive molecules. These cargos are transferred to recipient cells by endocytosis or binding to proteins on the receptor surface to mediate intercellular communication between different cell types and affect the functions of the recipient cells. Exosomes play an important role in the regulation of the immune system. Exosomes derived from MSCs (MSC-exos) carry immunomodulatory effectors or transmit active signal molecules to regulate the biological activities of immune cells and thus mediating immune suppression, especially on macrophages and T cells. Mitochondria and autophagy-related pathways are also associated with MSC-exos immunosuppressive effects. Graphical Abstract.
Collapse
Affiliation(s)
- Xiaoli Qian
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Nan An
- College of Clinical Medicine, Jilin University, Changchun, China
| | - Yifan Ren
- College of Clinical Medicine, Jilin University, Changchun, China
| | - Chenxin Yang
- College of Clinical Medicine, Jilin University, Changchun, China
| | - Xiaoling Zhang
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, The First Hospital of Jilin University, Changchun, China.
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, China.
| | - Lisha Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China.
| |
Collapse
|
7
|
Yan W, Li T, Yin T, Hou Z, Qu K, Wang N, Durkan C, Dong L, Qiu J, Gregersen H, Wang G. M2 macrophage-derived exosomes promote the c-KIT phenotype of vascular smooth muscle cells during vascular tissue repair after intravascular stent implantation. Theranostics 2020; 10:10712-10728. [PMID: 32929376 PMCID: PMC7482821 DOI: 10.7150/thno.46143] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 08/13/2020] [Indexed: 12/13/2022] Open
Abstract
Rationale: For intravascular stent implantation to be successful, the processes of vascular tissue repair and therapy are considered to be critical. However, the mechanisms underlying the eventual fate of vascular smooth muscle cells (VSMCs) during vascular tissue repair remains elusive. In this study, we hypothesized that M2 macrophage-derived exosomes to mediate cell-to-cell crosstalk and induce dedifferentiation phenotypes in VSMCs. Methods: In vivo, 316L bare metal stents (BMS) were implanted from the left iliac artery into the abdominal aorta of 12-week-old male Sprague-Dawley (SD) rats for 7 and 28 days. Hematoxylin and eosin (HE) were used to stain the neointimal lesions. En-face immunofluorescence staining of smooth muscle 22 alpha (SM22α) and CD68 showed the rat aorta smooth muscle cells (RASMCs) and macrophages. Immunohistochemical staining of total galactose-specific lectin 3 (MAC-2) and total chitinase 3-like 3 (YM-1) showed the total macrophages and M2 macrophages. In vitro, exosomes derived from IL-4+IL-13-treated macrophages (M2Es) were isolated by ultracentrifugation and characterized based on their specific morphology. Ki-67 staining was conducted to assess the effects of the M2Es on the proliferation of RASMCs. An atomic force microscope (AFM) was used to detect the stiffness of the VSMCs. GW4869 was used to inhibit exosome release. RNA-seq was performed to determine the mRNA profiles of the RASMCs and M2Es-treated RASMCs. Quantitative real-time PCR (qRT-PCR) analysis was conducted to detect the expression levels of the mRNAs. Western blotting was used to detect the candidate protein expression levels. T-5224 was used to inhibit the DNA binding activity of AP-1 in RASMCs. Results: M2Es promote c-KIT expression and softening of nearby VSMCs, hence accelerating the vascular tissue repair process. VSMCs co-cultured in vitro with M2 macrophages presented an increased capacity for de-differentiation and softening, which was exosome dependent. In addition, the isolated M2Es helped to promote VSMC dedifferentiation and softening. Furthermore, the M2Es enhanced vascular tissue repair potency by upregulation of VSMCs c-KIT expression via activation of the c-Jun/activator protein 1 (AP-1) signaling pathway. Conclusions: The findings of this study emphasize the prominent role of M2Es during VSMC dedifferentiation and vascular tissue repair via activation of the c-Jun/AP-1 signaling pathway, which has a profound impact on the therapeutic strategies of coronary stenting techniques.
Collapse
|
8
|
Lignelli E, Palumbo F, Myti D, Morty RE. Recent advances in our understanding of the mechanisms of lung alveolarization and bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2019; 317:L832-L887. [PMID: 31596603 DOI: 10.1152/ajplung.00369.2019] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is the most common cause of morbidity and mortality in preterm infants. A key histopathological feature of BPD is stunted late lung development, where the process of alveolarization-the generation of alveolar gas exchange units-is impeded, through mechanisms that remain largely unclear. As such, there is interest in the clarification both of the pathomechanisms at play in affected lungs, and the mechanisms of de novo alveoli generation in healthy, developing lungs. A better understanding of normal and pathological alveolarization might reveal opportunities for improved medical management of affected infants. Furthermore, disturbances to the alveolar architecture are a key histopathological feature of several adult chronic lung diseases, including emphysema and fibrosis, and it is envisaged that knowledge about the mechanisms of alveologenesis might facilitate regeneration of healthy lung parenchyma in affected patients. To this end, recent efforts have interrogated clinical data, developed new-and refined existing-in vivo and in vitro models of BPD, have applied new microscopic and radiographic approaches, and have developed advanced cell-culture approaches, including organoid generation. Advances have also been made in the development of other methodologies, including single-cell analysis, metabolomics, lipidomics, and proteomics, as well as the generation and use of complex mouse genetics tools. The objective of this review is to present advances made in our understanding of the mechanisms of lung alveolarization and BPD over the period 1 January 2017-30 June 2019, a period that spans the 50th anniversary of the original clinical description of BPD in preterm infants.
Collapse
Affiliation(s)
- Ettore Lignelli
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| | - Francesco Palumbo
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| | - Despoina Myti
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| | - Rory E Morty
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, member of the German Center for Lung Research, Giessen, Germany
| |
Collapse
|
9
|
Naeem A, Ahmed I, Silveyra P. Bronchopulmonary Dysplasia: An Update on Experimental Therapeutics. EUROPEAN MEDICAL JOURNAL 2019. [DOI: 10.33590/emj/10313109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic inflammatory lung disease that affects thousands of newborns and infants every year. Although it is accepted that BPD results from lung damage and inflammation triggered by mechanical ventilation and hyperoxia, the causes and molecular events leading to lung damage and arrested development remain unknown. While recent advances in neonatal care have improved the survival of very low-weight infants, the rates of BPD have not improved accordingly. This is mainly due to our limited understanding of the disease’s pathogenesis and the effective therapeutic options available. Current therapeutics for BPD involve ventilation management, steroid treatment, and administration of various agents, such as pulmonary surfactant, caffeine, vitamin A, nitric oxide, and stem cells. However, the efficacy of these agents in preventing and ameliorating BPD symptoms varies depending on the populations studied and the disease stage. As the field moves towards personalised therapeutic approaches, this review summarises clinical and experimental studies conducted in various models, aiming to increase understanding of the cellular and molecular mechanisms by which these agents can prevent or treat BPD. Due to the increasing number of extremely premature infants, it is imperative that we continue to work towards understanding the mechanisms of BPD pathogenesis and generating more effective therapeutic options.
Collapse
Affiliation(s)
- Anika Naeem
- Pulmonary Immunology and Physiology Laboratory (PIP), Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Imtiaz Ahmed
- Pulmonary Immunology and Physiology Laboratory (PIP), Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Patricia Silveyra
- Pulmonary Immunology and Physiology Laboratory (PIP), Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| |
Collapse
|
10
|
Braun RK, Chetty C, Balasubramaniam V, Centanni R, Haraldsdottir K, Hematti P, Eldridge MW. Intraperitoneal injection of MSC-derived exosomes prevent experimental bronchopulmonary dysplasia. Biochem Biophys Res Commun 2018; 503:2653-2658. [PMID: 30093115 DOI: 10.1016/j.bbrc.2018.08.019] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 08/02/2018] [Indexed: 12/13/2022]
Abstract
Mesenchymal stromal cell (MSC) derived exosomes mediate tissue protection and regeneration in many injuries and diseases by modulating cell protein production, protecting from apoptosis, inhibiting inflammation, and increasing angiogenesis. In the present study, daily intraperitoneal injection of MSC-derived exosomes protected alveolarization and angiogenesis in a newborn rat model of bronchopulmonary dysplasia (BPD) induced by 14 days of neonatal hyperoxia exposure (85% O2). Exosome treatment during hyperoxia prevented disruption of alveolar growth, increased small blood vessel number, and inhibited right heart hypertrophy at P14, P21, and P56. In vitro, exosomes significantly increased tube-like network formation by HUVEC, in part through a VEGF mediated mechanism. In summary, daily intraperitoneal injection of exosomes increased blood vessel number and size in the lung through pro-angiogenic mechanisms. MSC-derived exosomes therefore have both anti-inflammatory and pro-angiogenic mechanism to protect the lung from hyperoxia induced lung and heart disease associated with BPD.
Collapse
Affiliation(s)
- Rudolf K Braun
- Department of Pediatrics, University of Wisconsin Madison, Madison, WI, USA.
| | - Chandramu Chetty
- Department of Pediatrics, University of Wisconsin Madison, Madison, WI, USA
| | | | - Ryan Centanni
- Department of Pediatrics, University of Wisconsin Madison, Madison, WI, USA
| | | | - Peiman Hematti
- Department of Medicine, University of Wisconsin Madison, Madison, WI, USA; Carbone Cancer Center, University of Wisconsin Madison, Madison, WI, USA
| | - Marlowe W Eldridge
- Department of Pediatrics, University of Wisconsin Madison, Madison, WI, USA.
| |
Collapse
|
11
|
Chaubey S, Thueson S, Ponnalagu D, Alam MA, Gheorghe CP, Aghai Z, Singh H, Bhandari V. Early gestational mesenchymal stem cell secretome attenuates experimental bronchopulmonary dysplasia in part via exosome-associated factor TSG-6. Stem Cell Res Ther 2018; 9:173. [PMID: 29941022 PMCID: PMC6019224 DOI: 10.1186/s13287-018-0903-4] [Citation(s) in RCA: 143] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 04/25/2018] [Accepted: 05/13/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are promising tools for the treatment of human lung disease and other pathologies relevant to newborn medicine. Recent studies have established MSC exosomes (EXO), as one of the main therapeutic vectors of MSCs in mouse models of multifactorial chronic lung disease of preterm infants, bronchopulmonary dysplasia (BPD). However, the mechanisms underlying MSC-EXO therapeutic action are not completely understood. Using a neonatal mouse model of human BPD, we evaluated the therapeutic efficiency of early gestational age (GA) human umbilical cord (hUC)-derived MSC EXO fraction and its exosomal factor, tumor necrosis factor alpha-stimulated gene-6 (TSG-6). METHODS Conditioned media (CM) and EXO fractions were isolated from 25 and 30 weeks GA hUC-MSC cultures grown in serum-free media (SFM) for 24 h. Newborn mice were exposed to hyperoxia (> 95% oxygen) and were given intraperitoneal injections of MSC-CM or MSC-CM EXO fractions at postnatal (PN) day 2 and PN4. They were then returned to room air until PN14 (in a mouse model of severe BPD). The treatment regime was followed with (rh)TSG-6, TSG-6-neutralizing antibody (NAb), TSG-6 (si)RNA-transfected MSC-CM EXO and their appropriate controls. Echocardiography was done at PN14 followed by harvesting of lung, heart and brain for assessment of pathology parameters. RESULTS Systemic administration of CM or EXO in the neonatal BPD mouse model resulted in robust improvement in lung, cardiac and brain pathology. Hyperoxia-exposed BPD mice exhibited pulmonary inflammation accompanied by alveolar-capillary leakage, increased chord length, and alveolar simplification, which was ameliorated by MSC CM/EXO treatment. Pulmonary hypertension and right ventricular hypertrophy was also corrected. Cell death in brain was decreased and the hypomyelination reversed. Importantly, we detected TSG-6, an immunomodulatory glycoprotein, in EXO. Administration of TSG-6 attenuated BPD and its associated pathologies, in lung, heart and brain. Knockdown of TSG-6 by NAb or by siRNA in EXO abrogated the therapeutic effects of EXO, suggesting TSG-6 as an important therapeutic molecule. CONCLUSIONS Preterm hUC-derived MSC-CM EXO alleviates hyperoxia-induced BPD and its associated pathologies, in part, via exosomal factor TSG-6. The work indicates early systemic intervention with TSG-6 as a robust option for cell-free therapy, particularly for treating BPD.
Collapse
Affiliation(s)
- Sushma Chaubey
- Section of Neonatal-Perinatal Medicine, Department of Pediatrics, Drexel University College of Medicine, 245 N 15th Street, Philadelphia, PA, 19102, USA
| | - Sam Thueson
- Section of Neonatal-Perinatal Medicine, Department of Pediatrics, Drexel University College of Medicine, 245 N 15th Street, Philadelphia, PA, 19102, USA
| | - Devasena Ponnalagu
- Department of Pharmacology & Physiology, Drexel University College of Medicine, 245 N 15th Street, Philadelphia, PA, 19102, USA
| | - Mohammad Afaque Alam
- Section of Neonatal-Perinatal Medicine, Department of Pediatrics, Drexel University College of Medicine, 245 N 15th Street, Philadelphia, PA, 19102, USA
| | - Ciprian P Gheorghe
- Department of Obstetrics and Gynecology, Loma Linda University School of Medicine, 11370 Anderson Street, Loma Linda, CA, 92354, USA
| | - Zubair Aghai
- Divison of Neonatology, Department of Pediatrics, Thomas Jefferson University Hospital, 132S, 10th Street, Philadelphia, PA, 19107, USA
| | - Harpreet Singh
- Department of Pharmacology & Physiology, Drexel University College of Medicine, 245 N 15th Street, Philadelphia, PA, 19102, USA.,Department of Medicine, Division of Cardiology, Drexel University College of Medicine, 245 N 15th Street, Philadelphia, PA, 19102, USA
| | - Vineet Bhandari
- Section of Neonatal-Perinatal Medicine, Department of Pediatrics, Drexel University College of Medicine, 245 N 15th Street, Philadelphia, PA, 19102, USA.
| |
Collapse
|
12
|
Yuan Y, Du W, Liu J, Ma W, Zhang L, Du Z, Cai B. Stem Cell-Derived Exosome in Cardiovascular Diseases: Macro Roles of Micro Particles. Front Pharmacol 2018; 9:547. [PMID: 29904347 PMCID: PMC5991072 DOI: 10.3389/fphar.2018.00547] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 05/08/2018] [Indexed: 12/15/2022] Open
Abstract
The stem cell-based therapy has emerged as the promising therapeutic strategies for cardiovascular diseases (CVDs). Recently, increasing evidence suggest stem cell-derived active exosomes are important communicators among cells in the heart via delivering specific substances to the adjacent/distant target cells. These exosomes and their contents such as certain proteins, miRNAs and lncRNAs exhibit huge beneficial effects on preventing heart damage and promoting cardiac repair. More importantly, stem cell-derived exosomes are more effective and safer than stem cell transplantation. Therefore, administration of stem cell-derived exosomes will expectantly be an alternative stem cell-based therapy for the treatment of CVDs. Furthermore, modification of stem cell-derived exosomes or artificial synthesis of exosomes will be the new therapeutic tools for CVDs in the future. In addition, stem cell-derived exosomes also have been implicated in the diagnosis and prognosis of CVDs. In this review, we summarize the current advances of stem cell-derived exosome-based treatment and prognosis for CVDs, including their potential benefits, underlying mechanisms and limitations, which will provide novel insights of exosomes as a new tool in clinical therapeutic translation in the future.
Collapse
Affiliation(s)
- Ye Yuan
- Department of Clinical Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Weijie Du
- Department of Clinical Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Jiaqi Liu
- Department of Pharmacology, College of Pharmacy, Mudanjiang Medical University, Mudanjiang, China
| | - Wenya Ma
- Department of Clinical Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Lai Zhang
- Department of Clinical Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Zhimin Du
- Department of Clinical Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Benzhi Cai
- Department of Clinical Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| |
Collapse
|