1
|
Wan R, Liu Y, Yan J, Lin J. Cell therapy: A beacon of hope in the battle against pulmonary fibrosis. FASEB J 2025; 39:e70356. [PMID: 39873972 DOI: 10.1096/fj.202402790r] [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: 11/10/2024] [Revised: 12/28/2024] [Accepted: 01/15/2025] [Indexed: 01/30/2025]
Abstract
Pulmonary fibrosis (PF) is a chronic and progressive interstitial lung disease characterized by abnormal activation of myofibroblasts and pathological remodeling of the extracellular matrix, with a poor prognosis and limited treatment options. Lung transplantation is currently the only approach that can extend the life expectancy of patients; however, its applicability is severely restricted due to donor shortages and patient-specific limitations. Therefore, the search for novel therapeutic strategies is imperative. In recent years, stem cells have shown great promise in the field of regenerative medicine due to their self-renewal capacity and multidirectional differentiation potential, and a growing body of literature supports the efficacy of stem cell therapy in PF treatment. This paper systematically summarizes the research progress of various stem cell types in the treatment of PF. Furthermore, it discusses the primary methods and clinical outcomes of stem cell therapy in PF, based on both preclinical and clinical data. Finally, the current challenges and key factors to consider in stem cell therapy for PF are objectively analyzed, and future directions for improving this therapy are proposed, providing new insights and references for the clinical treatment of PF patients.
Collapse
Affiliation(s)
- Ruyan Wan
- Stem Cell and Biotherapy Technology Research Center, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
| | - Yanli Liu
- Stem Cell and Biotherapy Technology Research Center, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
| | - Jingwen Yan
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Juntang Lin
- Stem Cell and Biotherapy Technology Research Center, School of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Biomedical Engineering, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
2
|
Bondeelle L, Clément S, Bergeron A, Tapparel C. Lung stem cells and respiratory epithelial chimerism in transplantation. Eur Respir Rev 2025; 34:240146. [PMID: 39971397 PMCID: PMC11836672 DOI: 10.1183/16000617.0146-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 11/21/2024] [Indexed: 02/21/2025] Open
Abstract
Stem cells are capable of self-renewal and differentiation into specialised types. They range from totipotent cells to multipotent or somatic stem cells and ultimately to unipotent cells. Some adult multipotent stem cells can have the potential to regenerate and colonise diverse tissues. The respiratory airways and lung mucosa, exposed to ambient air, perform vital roles for all human tissues and organs. They serve as barriers against airborne threats and are essential for tissue oxygenation. Despite low steady-state turnover, lungs are vulnerable to injuries and diseases from environmental exposure. Lung stem cells are crucial due to their regenerative potential and ability to replace damaged cells. Lung repair with extrapulmonary stem cells can occur, leading to the coexistence of respiratory cells with different genetic origins, a phenomenon known as airway epithelial chimerism. The impact of such chimerism in lung repair and disease is actively studied. This review explores different stem cell types, focusing on pulmonary stem cells. It discusses airway epithelium models derived from stem cells for studying lung diseases and examines lung chimerism, particularly in lung transplantation and haematopoietic stem cell transplantation, highlighting its significance in understanding tissue repair and chimerism-mediated repair processes in lung pathology.
Collapse
Affiliation(s)
- Louise Bondeelle
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Sophie Clément
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
| | - Anne Bergeron
- Pneumology Department, Geneva University Hospitals, Geneva, Switzerland
- Co-last author
| | - Caroline Tapparel
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
- Co-last author
| |
Collapse
|
3
|
Liang J, Dai W, Xue S, Wu F, Cui E, Pan R. Recent progress in mesenchymal stem cell-based therapy for acute lung injury. Cell Tissue Bank 2024; 25:677-684. [PMID: 38466563 DOI: 10.1007/s10561-024-10129-0] [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: 06/07/2022] [Accepted: 01/24/2024] [Indexed: 03/13/2024]
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are life-threatening diseases in critically ill patients. Although pathophysiology of ALI/ARDS has been investigated in many studies, effective therapeutic strategies are still limited. Mesenchymal stem cell (MSC)-based therapy is emerging as a promising therapeutic intervention for patients with ALI. During the last two decades, researchers have focused on the efficacy and mechanism of MSC application in ALI animal models. MSC derived from variant resources exhibited therapeutic effects in preclinical studies of ALI with different mechanisms. Based on this, clinical studies on MSC treatment in ALI/ARDS has been tried recently, especially in COVID-19 caused lung injury. Emerging clinical trials of MSCs in treating COVID-19-related conditions have been registered in past two years. The advantages and potential of MSCs in the defense against COVID-19-related ALI or ARDS have been confirmed. This review provides a brief overview of recent research progress in MSC-based therapies in preclinical study and clinical trials in ALI treatment, as well as the underlying mechanisms.
Collapse
Affiliation(s)
- Jinfeng Liang
- Zhejiang Center for Drug and Cosmetic Evaluation, Hangzhou, China
| | - Weiyou Dai
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Shihang Xue
- Xiangshan First People's Hospital Medical and Health Group, Ningbo, China
| | - Feifei Wu
- Key Laboratory of Cell-Based Drug and Applied Technology Development in Zhejiang Province, Hangzhou, China
- Institute for Cell-Based Drug Development of Zhejiang Province, S-Evans Biosciences, No.181 Wuchang Road, Hangzhou, 311122, Zhejiang, People's Republic of China
| | - Enhai Cui
- Huzhou Central Hospital, Zhejiang University Huzhou Hospital, Huzhou, 313000, People's Republic of China.
| | - Ruolang Pan
- Key Laboratory of Cell-Based Drug and Applied Technology Development in Zhejiang Province, Hangzhou, China.
- Institute for Cell-Based Drug Development of Zhejiang Province, S-Evans Biosciences, No.181 Wuchang Road, Hangzhou, 311122, Zhejiang, People's Republic of China.
| |
Collapse
|
4
|
Diwan R, Bhatt HN, Beaven E, Nurunnabi M. Emerging delivery approaches for targeted pulmonary fibrosis treatment. Adv Drug Deliv Rev 2024; 204:115147. [PMID: 38065244 PMCID: PMC10787600 DOI: 10.1016/j.addr.2023.115147] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 11/02/2023] [Accepted: 11/29/2023] [Indexed: 01/01/2024]
Abstract
Pulmonary fibrosis (PF) is a progressive, and life-threatening interstitial lung disease which causes scarring in the lung parenchyma and thereby affects architecture and functioning of lung. It is an irreversible damage to lung functioning which is related to epithelial cell injury, immense accumulation of immune cells and inflammatory cytokines, and irregular recruitment of extracellular matrix. The inflammatory cytokines trigger the differentiation of fibroblasts into activated fibroblasts, also known as myofibroblasts, which further increase the production and deposition of collagen at the injury sites in the lung. Despite the significant morbidity and mortality associated with PF, there is no available treatment that efficiently and effectively treats the disease by reversing their underlying pathologies. In recent years, many therapeutic regimens, for instance, rho kinase inhibitors, Smad signaling pathway inhibitors, p38, BCL-xL/ BCL-2 and JNK pathway inhibitors, have been found to be potent and effective in treating PF, in preclinical stages. However, due to non-selectivity and non-specificity, the therapeutic molecules also result in toxicity mediated severe side effects. Hence, this review demonstrates recent advances on PF pathology, mechanism and targets related to PF, development of various drug delivery systems based on small molecules, RNAs, oligonucleotides, peptides, antibodies, exosomes, and stem cells for the treatment of PF and the progress of various therapeutic treatments in clinical trials to advance PF treatment.
Collapse
Affiliation(s)
- Rimpy Diwan
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX 79902, United States; Department of Biomedical Engineering, College of Engineering, The University of Texas El Paso, El Paso, TX 79968, United States
| | - Himanshu N Bhatt
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX 79902, United States; Department of Biomedical Engineering, College of Engineering, The University of Texas El Paso, El Paso, TX 79968, United States
| | - Elfa Beaven
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX 79902, United States; Department of Biomedical Engineering, College of Engineering, The University of Texas El Paso, El Paso, TX 79968, United States
| | - Md Nurunnabi
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX 79902, United States; Department of Biomedical Engineering, College of Engineering, The University of Texas El Paso, El Paso, TX 79968, United States; The Border Biomedical Research Center, The University of Texas El Paso, El Paso, TX 79968, United States.
| |
Collapse
|
5
|
Zhang X, Xue M, Liu A, Qiu H, Guo F. Activation of Wnt/β‑Catenin‑p130/E2F4 promotes the differentiation of bone marrow‑derived mesenchymal stem cells into type II alveolar epithelial cells through cell cycle arrest. Exp Ther Med 2023; 26:330. [PMID: 37346406 PMCID: PMC10280314 DOI: 10.3892/etm.2023.12029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 04/18/2023] [Indexed: 06/23/2023] Open
Abstract
The results of our previous study demonstrated that activation of the Wnt/β-catenin pathway increased the differentiation of mesenchymal stem cells (MSCs) into type II alveolar epithelial (AT II) cells; however, the specific mechanisms remain unclear. The present study aimed to evaluate the role of Wnt/β-catenin-p130/E2F transcription factor 4 (E2F4) in regulating the differentiation of mouse MSCs (mMSCs) into AT II cells, and to determine the specific mechanisms. mMSCs with p130 or E2F4 overexpression were constructed using lentiviral vectors. Differentiation of mMSCs into AT II cells was promoted using a modified coculture system with murine lung epithelial-12 cells incubated in small airway growth medium for 7-14 days. The differentiation efficiency was detected using immunofluorescence, western blot analysis and transmission electron microscopy. To detect the association between the canonical Wnt pathway and p130/E2F4, 4 mmol/l lithium chloride (LiCl) or 200 ng/ml Dickkopf-related protein 1 (DKK-1) was also added to the coculture system. Following differentiation, the cell cycle of mMSCs was evaluated using flow cytometry. The results of the present study demonstrated that surfactant protein C (SP-C) protein expression was higher in the p130 overexpression (MSC-p130) and E2F4 overexpression (MSC-E2F4) groups compared with the normal control mMSCs group following differentiation into AT II cells. Similar results for SP-C protein expression and lamellar body-like structures were also observed using immunofluorescence analysis and electron microscopy. Following the addition of LiCl into the coculture system for activation of the Wnt/β-catenin signaling pathway, phosphorylated (p)-p130/p130 was slightly decreased at 7 days and E2F4 was increased both at 7 and 14 days in mMSCs. Furthermore, the p-p130/p130 ratio was significantly increased at 14 days and E2F4 was decreased both at 7 and 14 days following DKK-1-mediated inhibition of the Wnt pathway. The results of the present study demonstrated that the numbers of cells in G1 and S phases were increased following activation of the Wnt pathway and decreased following Wnt pathway inhibition. However, the number of cells in G1 phase was increased following the differentiation of mMSCs overexpressing p130 or E2F4. Therefore, the results of the present study revealed that the canonical Wnt signaling pathway may affect the differentiation of MSCs into AT II cells via regulation of downstream p130/E2F4. The specific mechanisms may be associated with G1 phase extension in the cell cycle of MSCs.
Collapse
Affiliation(s)
- Xiwen Zhang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Ming Xue
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Airan Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Haibo Qiu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Fengmei Guo
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| |
Collapse
|
6
|
Quesenberry PJ, Wen S, Goldberg LR, Dooner MS. The universal stem cell. Leukemia 2022; 36:2784-2792. [PMID: 36307485 PMCID: PMC9712109 DOI: 10.1038/s41375-022-01715-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/26/2022] [Accepted: 09/22/2022] [Indexed: 11/08/2022]
Abstract
Current dogma is that there exists a hematopoietic pluripotent stem cell, resident in the marrow, which is quiescent, but with tremendous proliferative and differentiative potential. Furthermore, the hematopoietic system is essentially hierarchical with progressive differentiation from the pluripotent stem cells to different classes of hematopoietic cells. However, results summarized here indicate that the marrow pluripotent hematopoietic stem cell is actively cycling and thus continually changing phenotype. As it progresses through cell cycle differentiation potential changes as illustrated by sequential changes in surface expression of B220 and GR-1 epitopes. Further data indicated that the potential of purified hematopoietic stem cells extends to multiple other non-hematopoietic cells. It appears that marrow stem cells will give rise to epithelial pulmonary cells at certain points in cell cycle. Thus, it appears that the marrow "hematopoietic" stem cell is also a stem cell for other non-hematopoietic tissues. These observations give rise to the concept of a universal stem cell. The marrow stem cell is not limited to hematopoiesis and its differentiation potential continually changes as it transits cell cycle. Thus, there is a universal stem cell in the marrow which alters its differentiation potential as it progresses through cell cycle. This potential is expressed when it resides in tissues compatible with its differentiation potential, at a particular point in cell cycle transit, or when it interacts with vesicles from that tissue.
Collapse
Affiliation(s)
- Peter J Quesenberry
- Division of Hematology/Oncology, Brown University, Rhode Island Hospital, Providence, RI, 02903, USA.
| | - Sicheng Wen
- Division of Hematology/Oncology, Brown University, Rhode Island Hospital, Providence, RI, 02903, USA
| | - Laura R Goldberg
- Division of Hematology/Oncology, Brown University, Rhode Island Hospital, Providence, RI, 02903, USA
- Division of Hematology, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Mark S Dooner
- Division of Hematology/Oncology, Brown University, Rhode Island Hospital, Providence, RI, 02903, USA
| |
Collapse
|
7
|
Xu L, Zhu Y, Li C, Wang Q, Ma L, Wang J, Zhang S. Small extracellular vesicles derived from Nrf2-overexpressing human amniotic mesenchymal stem cells protect against lipopolysaccharide-induced acute lung injury by inhibiting NLRP3. Biol Direct 2022; 17:35. [PMID: 36447296 PMCID: PMC9706911 DOI: 10.1186/s13062-022-00351-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 11/21/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Acute lung injury (ALI) is a major cause of respiratory failure in critically ill patients that results in significant morbidity and mortality. Recent studies indicate that cell-based therapies may be beneficial in the treatment of ALI. We recently demonstrated that Nrf2-overexpressing human amniotic mesenchymal stem cells (hAMSCs) reduce lung injury, fibrosis and inflammation in lipopolysaccharide (LPS)-challenged mice. Here we tested whether small extracellular vesicles (sEVs) derived from Nrf2-overexpressing hAMSCs (Nrf2-sEVs) could protect against ALI. sEVs were isolated from hAMSCs that overexpressed (Nrf2-sEVs) or silenced (siNrf2-sEVs) Nrf2. We examined the effects of sEVs treatment on lung inflammation in a mouse model of ALI, where LPS was administered intratracheally to mice, and lung tissues and bronchoalveolar lavage fluid (BALF) were analyzed 24 h later. METHODS Histological analysis, immunofluorescence microscopy, western blotting, RT-PCR and ELISA were used to measure the inflammatory response in the lungs and BALF. RESULTS We found that sEVs from hAMSCs are protective in ALI and that Nrf2 overexpression promotes protection against lung disease. Nrf2-sEVs significantly reduced lung injury in LPS-challenged mice, which was associated with decreased apoptosis, reduced infiltration of neutrophils and macrophages, and inhibition of pro-inflammatory cytokine expression. We further show that Nrf2-sEVs act by inhibiting the activation of the NLRP3 inflammasome and promoting the polarization of M2 macrophages. CONCLUSION Our data show that overexpression of Nrf2 protects against LPS-induced lung injury, and indicate that a novel therapeutic strategy using Nrf2-sEVs may be beneficial against ALI.
Collapse
Affiliation(s)
- Lijuan Xu
- grid.24516.340000000123704535Department of Critical Care Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 7th Floor, Building 1, No. 301 Middle Yanchang Road, Jing’an District, Shanghai, 200072 China
| | - Yunlou Zhu
- grid.24516.340000000123704535Department of Critical Care Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 7th Floor, Building 1, No. 301 Middle Yanchang Road, Jing’an District, Shanghai, 200072 China
| | - Congye Li
- grid.24516.340000000123704535Department of Critical Care Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 7th Floor, Building 1, No. 301 Middle Yanchang Road, Jing’an District, Shanghai, 200072 China
| | - Qixing Wang
- grid.24516.340000000123704535Department of Critical Care Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 7th Floor, Building 1, No. 301 Middle Yanchang Road, Jing’an District, Shanghai, 200072 China
| | - Lijie Ma
- grid.24516.340000000123704535Department of Critical Care Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 7th Floor, Building 1, No. 301 Middle Yanchang Road, Jing’an District, Shanghai, 200072 China
| | - Junjie Wang
- grid.24516.340000000123704535Department of Critical Care Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 7th Floor, Building 1, No. 301 Middle Yanchang Road, Jing’an District, Shanghai, 200072 China
| | - Shouqin Zhang
- grid.24516.340000000123704535Department of Critical Care Medicine, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, 7th Floor, Building 1, No. 301 Middle Yanchang Road, Jing’an District, Shanghai, 200072 China
| |
Collapse
|
8
|
Zhuang Y, Yang W, Zhang L, Fan C, Qiu L, Zhao Y, Chen B, Chen Y, Shen H, Dai J. A novel leptin receptor binding peptide tethered-collagen scaffold promotes lung injury repair. Biomaterials 2022; 291:121884. [DOI: 10.1016/j.biomaterials.2022.121884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 10/10/2022] [Accepted: 10/23/2022] [Indexed: 11/06/2022]
|
9
|
Abstract
Pulmonary fibrosis (PF) is a chronic and relentlessly progressive interstitial lung disease in which the accumulation of fibroblasts and extracellular matrix (ECM) induces the destruction of normal alveolar structures, ultimately leading to respiratory failure. Patients with advanced PF are unable to perform physical labor and often have concomitant cough and dyspnea, which markedly impair their quality of life. However, there is a paucity of available pharmacological therapies, and to date, lung transplantation remains the only possible treatment for patients suffering from end-stage PF; moreover, the complexity of transplantation surgery and the paucity of donors greatly restrict the application of this treatment. Therefore, there is a pressing need for alternative therapeutic strategies for this complex disease. Due to their capacity for pluripotency and paracrine actions, stem cells are promising therapeutic agents for the treatment of interstitial lung disease, and an extensive body of literature supports the therapeutic efficacy of stem cells in lung fibrosis. Although stem cell transplantation may play an important role in the treatment of PF, some key issues, such as safety and therapeutic efficacy, remain to be resolved. In this review, we summarize recent preclinical and clinical studies on the stem cell-mediated regeneration of fibrotic lungs and present an analysis of concerning issues related to stem cell therapy to guide therapeutic development for this complex disease.
Collapse
|
10
|
Zhang Y, Yang S, Qiu Z, Huang L, Huang L, Liang Y, Liu X, Wang M, Zhou B. Pyrogallol enhances therapeutic effect of human umbilical cord mesenchymal stem cells against LPS-mediated inflammation and lung injury via activation of Nrf2/HO-1 signaling. Free Radic Biol Med 2022; 191:66-81. [PMID: 36028178 DOI: 10.1016/j.freeradbiomed.2022.08.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/31/2022] [Accepted: 08/18/2022] [Indexed: 11/25/2022]
Abstract
The main challenges in clinical applications of mesenchymal stem cells (MSCs) are attributed to their heterogeneity. It is believed that preconditioning of MSCs with active compounds may enhance the expression of potentially therapeutic molecules and thus achieve stable and effective therapeutic outcomes. In the present study, we investigated the mechanism by which pyrogallol increased the therapeutic efficacy of human umbilical cord mesenchymal stem cells (hUCMSCs) against LPS-induced acute lung injury (ALI). hUCMSCs with pyrogallol treatment increased expression of HO-1 at both mRNA and protein levels, accompanied by Kelch-Like ECH-Associated Protein 1 (Keap1) degradation, and upregulation of the Nrf2 protein levels as well as nuclear translocation of Nrf2. Moreover, the modulation of Keap1 and Nrf2 as well as HO-1 upregulation by pyrogallol was reversed by pretreatment with N-acetylcysteine (NAC) and a P38 kinase inhibitor (SB203580). Whereas, NAC pretreatment abrogated pyrogallol-mediated activation of P38 kinase, indicating that pyrogallol-derived ROS led to P38 kinase activation, thus promoting Nrf2/HO-1 signaling. Additionally, we found that the induction of p62 by the pyrogallol-mediated ROS/P38/Nrf2 axis interacted with Keap1 and resulted in autophagic degradation of Keap1, which created a positive feedback loop to further release of Nrf2. Furthermore, the increased expression of HO-1 in pyrogallol-pretreated hUCMSCs led to enhanced inhibitory effects on LPS-mediated TLR4/P-P65 signaling in BEAS-2B cells, resulting in increasing suppression of LPS-indued expression of a series of pro-inflammatory mediators. Compared to untreated hUCMSCs, Sprague-Dawley (SD) rats with pyrogallol-primed hUCMSCs transplantation showed enhanced improvements in LPS-mediated lung pathological alterations, the increased lung index (lung/body ratio), apoptosis of epithelial cells, the activation of TLR4/NF-κB signaling as well as the release of pro-inflammatory mediators. Together, these results suggested that hUCMSCs with pyrogallol pretreatment enhanced the therapeutic efficacy of hUCMSCs, which may provide a promising therapeutic strategy to maximize the therapeutic efficacy of hUCMSC-based therapy for treating LPS-associated ALI.
Collapse
Affiliation(s)
- Yuehan Zhang
- Center of Stem Cell and Regenerative Medicine, The People's Hospital of Gaozhou, Gaozhou, 525200, China
| | - Sushan Yang
- Department of Clinical Laboratory, The People's Hospital of Gaozhou, Gaozhou, 525200, China
| | - Zhenhua Qiu
- Department of Clinical Laboratory, The People's Hospital of Gaozhou, Gaozhou, 525200, China
| | - Li Huang
- Center of Stem Cell and Regenerative Medicine, The People's Hospital of Gaozhou, Gaozhou, 525200, China
| | - Linyan Huang
- Department of Hematopathology, The People's Hospital of Gaozhou, Gaozhou, 525200, China
| | - Yueyun Liang
- Center of Stem Cell and Regenerative Medicine, The People's Hospital of Gaozhou, Gaozhou, 525200, China
| | - Xuanyu Liu
- Center of Stem Cell and Regenerative Medicine, The People's Hospital of Gaozhou, Gaozhou, 525200, China
| | - Maosheng Wang
- Center of Stem Cell and Regenerative Medicine, The People's Hospital of Gaozhou, Gaozhou, 525200, China.
| | - Beixian Zhou
- Center of Stem Cell and Regenerative Medicine, The People's Hospital of Gaozhou, Gaozhou, 525200, China; Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, 510315, Guangzhou, China.
| |
Collapse
|
11
|
Mesenchyme Stem Cell-Derived Conditioned Medium as a Potential Therapeutic Tool in Idiopathic Pulmonary Fibrosis. Biomedicines 2022; 10:biomedicines10092298. [PMID: 36140399 PMCID: PMC9496127 DOI: 10.3390/biomedicines10092298] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/04/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
Mesenchyme Stem Cells (MSCs) are the most used types of stem cells in regenerative medicine. Regenerative medicine is a rapidly emerging medicine section that creates new methods to regrow, restore, and replace diseased and damaged tissues, organs, and cells. Scholars have shown a positive correlation between MSCs-based therapies and successful treatment of diseases like cardiac ischemia, cartilage problems, bone diseases, diabetes, and even neurological disorders. Although MSCs have several varying features that make them unique, their immuno-regulatory effects in tissue repair emerge from their secretion of paracrine growth factors, exosomes, and cytokines. These cells secrete a secretome, which has regenerative and reparative properties that lead to injury amelioration, immune modulation, or fibrosis reduction. Recent studies have shown that the administration MCSs derived conditioned medium (MSCs-CM) in acute doses in humans is safe and well-tolerated. Studies from animal models and human clinical trials have also shown that they are efficacious tools in regenerative medicine. In this review, we will explore the therapeutic potential of MSCs-CM in pulmonary fibrosis, with further insight into the treatment of Idiopathic Pulmonary Fibrosis (IPF).
Collapse
|
12
|
Du Y, Jiao J, Cao A, Ji C, Li M, Ji C, Wu Y, Guo Y, Wang Y, Zhou J, Ren Y. Ultrasound-based radiomics for the evaluation of fetal rat lung maturity a non-invasive assessment method (Ultrasound-based radiomics in fetal rat lung). Prenat Diagn 2022; 42:1429-1437. [PMID: 36056747 DOI: 10.1002/pd.6229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 08/20/2022] [Accepted: 08/25/2022] [Indexed: 11/10/2022]
Abstract
OBJECTIVE To establish a classification model for the evaluation of rat fetal lung maturity (FLM) using radiomics technology. METHOD A total of 430 high-throughput features were extracted per fetal lung image from 134 fetal lung ultrasound images (four-cardiac-chamber views) of 67 Sprague-Dawley (SD) fetal rats with gestational age (GA) of 16-21 days. The detection of fetal lung tissues included histopathological staining and the expression of the surface protein (SP) SP-A, SP-B, and SP-C. A machine learning classification model was established by a support vector machine based on histopathological results to analyze the relationship between fetal lung texture characteristics and FLM. RESULTS The rat fetal lungs were divided into two groups: terminal sac period (SD1) and canalicular period (SD2). The mRNA transcription and protein expression level of SP-C protein were significantly higher in the SD1 group than in the SD2 group (P < 0.05). The diagnostic performance of the rat FLM classification model was measured as follows: area under the receiver operating characteristic curve (AUC), 0.93 (training set) and 0.89 (validation set); sensitivity, 89.26% (training set) and 87.10% (validation set); specificity, 85.87% (training set) and 79.17% (validation set); accuracy, 87.79% (training set) and 83.64% (validation set). CONCLUSION Ultrasound-based radiomics technology can be used to evaluate the FLM of rats, which lays a foundation for further research on this technology in human fetal lungs. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Yanran Du
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, No. 197, Rui Jin 2nd Road, Shanghai, 200025, China
| | - Jing Jiao
- Department of Electronic Engineering, Fudan University, No. 220, Handan Road, Yangpu District, Shanghai, 200433, China.,Key Laboratory of Medical Imaging, Computing and Computer-Assisted Intervention, Shanghai, China
| | - Aili Cao
- Putuo Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No.164, Lanxi Road, Shanghai, 200062, China
| | - Chao Ji
- Putuo Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No.164, Lanxi Road, Shanghai, 200062, China
| | - Man Li
- Obstetrics and Gynecology Hospital of Fudan University, No.128, Shenyang Road, Shanghai, 200090, China
| | - Chenli Ji
- Putuo Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No.164, Lanxi Road, Shanghai, 200062, China
| | - Yang Wu
- Putuo Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, No.164, Lanxi Road, Shanghai, 200062, China
| | - Yi Guo
- Department of Electronic Engineering, Fudan University, No. 220, Handan Road, Yangpu District, Shanghai, 200433, China.,Key Laboratory of Medical Imaging, Computing and Computer-Assisted Intervention, Shanghai, China
| | - Yuanyuan Wang
- Department of Electronic Engineering, Fudan University, No. 220, Handan Road, Yangpu District, Shanghai, 200433, China.,Key Laboratory of Medical Imaging, Computing and Computer-Assisted Intervention, Shanghai, China
| | - Jianqiao Zhou
- Department of Ultrasound, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, No. 197, Rui Jin 2nd Road, Shanghai, 200025, China
| | - Yunyun Ren
- Obstetrics and Gynecology Hospital of Fudan University, No.128, Shenyang Road, Shanghai, 200090, China
| |
Collapse
|
13
|
Menzel-Severing J, Spaniol K, Groeber-Becker F, Geerling G. [Regenerative medicine for the corneal epithelium : Cell therapy from bench to bedside]. DIE OPHTHALMOLOGIE 2022; 119:891-901. [PMID: 35925345 DOI: 10.1007/s00347-022-01674-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/31/2022] [Indexed: 06/15/2023]
Abstract
In the case of thermal or caustic burns of the ocular surface, loss of limbal epithelial stem cells leads to compromised self-renewal of the corneal epithelium. This results in permanent loss of vision. In these situations, transplantation of cultured limbal epithelial cells on an amniotic membrane or fibrin gel as substrate (Holoclar®) can help to regenerate the corneal surface. The required cells are obtained from the healthy partner eye, if available. Adult stem cells from other parts of the body potentially serve as alternative cell sources: hair follicles, oral mucosa, mesenchymal stromal cells, or induced pluripotent stem cells (originally, e.g., skin fibroblasts). The reprogramming of such cells can be achieved with the help of transcription factors. In addition, work is being done on biosynthetic or synthetic matrices, which not only serve as substrate material for the transplantation but also support the functional properties of these cells (self-renewal, corneal epithelial-typical phenotype).
Collapse
Affiliation(s)
- Johannes Menzel-Severing
- Klinik für Augenheilkunde, Universitätsklinikum Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Deutschland.
| | - Kristina Spaniol
- Klinik für Augenheilkunde, Universitätsklinikum Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Deutschland
| | - Florian Groeber-Becker
- Translationszentrum Regenerative Therapien | TLZ-RT, Leitung In-vitro-Testsysteme, Fraunhofer-Institut für Silicatforschung ISC, Würzburg, Deutschland
| | - Gerd Geerling
- Klinik für Augenheilkunde, Universitätsklinikum Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Deutschland
| |
Collapse
|
14
|
Protease Activated Receptors: A Pathway to Boosting Mesenchymal Stromal Cell Therapeutic Efficacy in Acute Respiratory Distress Syndrome? Int J Mol Sci 2022; 23:ijms23031277. [PMID: 35163205 PMCID: PMC8836081 DOI: 10.3390/ijms23031277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/16/2022] [Accepted: 01/17/2022] [Indexed: 02/04/2023] Open
Abstract
Acute Respiratory Distress Syndrome is the most common cause of respiratory failure among critically ill patients, and its importance has been heightened during the COVID-19 pandemic. Even with the best supportive care, the mortality rate in the most severe cases is 40–50%, and the only pharmacological agent shown to be of possible benefit has been steroids. Mesenchymal stromal cells (MSCs) have been tested in several pre-clinical models of lung injury and been found to have significant therapeutic benefit related to: (a) potent immunomodulation; (b) secretion of epithelial and endothelial growth factors; and (c) augmentation of host defense to infection. Initial translational efforts have shown signs of promise, but the results have not yielded the anticipated outcomes. One potential reason is the relatively low survival of MSCs in inflammatory conditions as shown in several studies. Therefore, strategies to boost the survival of MSCs are needed to enhance their therapeutic effect. Protease-activated receptors (PARs) may represent one such possibility as they are G-protein coupled receptors expressed by MSCs and control several facets of cell behavior. This review summarizes some of the existing literature about PARs and MSCs and presents possible future areas of investigation in order to develop potential, PAR-modified MSCs with enhanced therapeutic efficiency.
Collapse
|
15
|
Behrend SJ, Giotopoulou GA, Spella M, Stathopoulos GT. A role for club cells in smoking-associated lung adenocarcinoma. Eur Respir Rev 2021; 30:30/162/210122. [PMID: 34670807 PMCID: PMC9488964 DOI: 10.1183/16000617.0122-2021] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 08/05/2021] [Indexed: 01/02/2023] Open
Abstract
The cellular origin of lung adenocarcinoma remains a focus of intense research efforts. The marked cellular heterogeneity and plasticity of the lungs, as well as the vast variety of molecular subtypes of lung adenocarcinomas perplex the field and account for the extensive variability of experimental results. While most experts would agree on the cellular origins of other types of thoracic tumours, great controversy exists on the tumour-initiating cells of lung adenocarcinoma, since this histologic subtype of lung cancer arises in the distal pulmonary regions where airways and alveoli converge, occurs in smokers as well as nonsmokers, is likely caused by various environmental agents, and is marked by vast molecular and pathologic heterogeneity. Alveolar type II, club, and their variant cells have all been implicated in lung adenocarcinoma progeny and the lineage hierarchies in the distal lung remain disputed. Here we review the relevant literature in this rapidly expanding field, including results from mouse models and human studies. In addition, we present a case for club cells as cells of origin of lung adenocarcinomas that arise in smokers. Multiple lung epithelial cells are targets of carcinogenic hits. Club cells are such cells that can metabolically activate tobacco pre-carcinogens, being thus positioned as cells of origin of lung adenocarcinomas in smokers.https://bit.ly/3iOshcy
Collapse
Affiliation(s)
- Sabine J Behrend
- Comprehensive Pneumology Center (CPC) and Institute for Lung Biology and Disease (iLBD); Helmholtz Center Munich-German Research Center for Environmental Health (HMGU) and Ludwig-Maximilian-University (LMU) Munich, Munich, Germany .,German Center for Lung Research (DZL), Giessen, Germany
| | - Georgia A Giotopoulou
- Comprehensive Pneumology Center (CPC) and Institute for Lung Biology and Disease (iLBD); Helmholtz Center Munich-German Research Center for Environmental Health (HMGU) and Ludwig-Maximilian-University (LMU) Munich, Munich, Germany.,German Center for Lung Research (DZL), Giessen, Germany
| | - Magda Spella
- Laboratory for Molecular Respiratory Carcinogenesis, Dept of Physiology, Faculty of Medicine, University of Patras, Patras, Greece
| | - Georgios T Stathopoulos
- Comprehensive Pneumology Center (CPC) and Institute for Lung Biology and Disease (iLBD); Helmholtz Center Munich-German Research Center for Environmental Health (HMGU) and Ludwig-Maximilian-University (LMU) Munich, Munich, Germany.,German Center for Lung Research (DZL), Giessen, Germany
| |
Collapse
|
16
|
Saeedi M, Nezhad MS, Mehranfar F, Golpour M, Esakandari MA, Rashmeie Z, Ghorbani M, Nasimi F, Hoseinian SN. Biological Aspects and Clinical Applications of Mesenchymal Stem Cells: Key Features You Need to be Aware of. Curr Pharm Biotechnol 2021; 22:200-215. [PMID: 32895040 DOI: 10.2174/1389201021666200907121530] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/01/2020] [Accepted: 08/03/2020] [Indexed: 11/22/2022]
Abstract
Mesenchymal Stem Cells (MSCs), a form of adult stem cells, are known to have a selfrenewing property and the potential to specialize into a multitude of cells and tissues such as adipocytes, cartilage cells, and fibroblasts. MSCs can migrate and home to the desired target zone where inflammation is present. The unique characteristics of MSCs in repairing, differentiation, regeneration, and the high capacity of immune modulation have attracted tremendous attention for exerting them in clinical purposes, as they contribute to the tissue regeneration process and anti-tumor activity. The MSCs-based treatment has demonstrated remarkable applicability towards various diseases such as heart and bone malignancies, and cancer cells. Importantly, genetically engineered MSCs, as a stateof- the-art therapeutic approach, could address some clinical hurdles by systemic secretion of cytokines and other agents with a short half-life and high toxicity. Therefore, understanding the biological aspects and the characteristics of MSCs is an imperative issue of concern. Herein, we provide an overview of the therapeutic application and the biological features of MSCs against different inflammatory diseases and cancer cells. We further shed light on MSCs' physiological interaction, such as migration, homing, and tissue repairing mechanisms in different healthy and inflamed tissues.
Collapse
Affiliation(s)
- Mohammad Saeedi
- Department of Laboratory Science, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Muhammad S Nezhad
- Stem Cells and Regenerative Medicine Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Mehranfar
- Department of Laboratory Science, Faculty of medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Mahdieh Golpour
- School of Paramedical Sciences, Semnan University of Medical Sciences, Sorkheh, Semnan, Iran
| | - Mohammad A Esakandari
- Student Research Committee, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Zahra Rashmeie
- Department of Laboratory Science, Faculty of medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Maryam Ghorbani
- Department of Laboratory Science, Faculty of medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Fatemeh Nasimi
- Department of Laboratory Science, Faculty of medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Seyed N Hoseinian
- Department of Laboratory Science, Faculty of medicine, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
17
|
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: 7] [Impact Index Per Article: 1.4] [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
|
18
|
Rangarajan S, Thannickal VJ. Remember Me? The Bone Marrow in Pulmonary Fibrosis. Am J Respir Crit Care Med 2020; 200:959-960. [PMID: 31206314 PMCID: PMC6794115 DOI: 10.1164/rccm.201906-1101ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Affiliation(s)
- Sunad Rangarajan
- Division of Pulmonary Sciences and Critical Care MedicineUniversity of Colorado Anschutz Medical CampusAurora, Coloradoand
| | - Victor J Thannickal
- Division of Pulmonary, Allergy and Critical Care MedicineUniversity of Alabama at BirminghamBirmingham, Alabama
| |
Collapse
|
19
|
Kumar P, Ciftci S, Barthes J, Knopf‐Marques H, Muller CB, Debry C, Vrana NE, Ghaemmaghami AM. A composite Gelatin/hyaluronic acid hydrogel as an ECM mimic for developing mesenchymal stem cell‐derived epithelial tissue patches. J Tissue Eng Regen Med 2019; 14:45-57. [DOI: 10.1002/term.2962] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 07/29/2019] [Accepted: 09/04/2019] [Indexed: 01/12/2023]
Affiliation(s)
- Pramod Kumar
- Immunology and Tissue Modelling Group, School of Life Science, Faculty of Medicine and Health SciencesUniversity of Nottingham Nottingham UK
| | - Sait Ciftci
- INSERM UMR 1121 Strasbourg France
- Service Oto‐Rhino‐LaryngologieHôpitaux Universitaires de Strasbourg Strasbourg France
| | - Julien Barthes
- INSERM UMR 1121 Strasbourg France
- Protip Medical Strasbourg France
| | - Helena Knopf‐Marques
- INSERM UMR 1121 Strasbourg France
- Faculté de Chirurgie DentaireUniversité de Strasbourg Strasbourg France
| | | | - Christian Debry
- INSERM UMR 1121 Strasbourg France
- Service Oto‐Rhino‐LaryngologieHôpitaux Universitaires de Strasbourg Strasbourg France
| | - Nihal E. Vrana
- INSERM UMR 1121 Strasbourg France
- Protip Medical Strasbourg France
| | - Amir M. Ghaemmaghami
- Immunology and Tissue Modelling Group, School of Life Science, Faculty of Medicine and Health SciencesUniversity of Nottingham Nottingham UK
| |
Collapse
|
20
|
Chen M, Huang Z, Bi H, Pan X, He J, He L, He X, Du J, Zhou K, Wang L, Wang Q, Guo X, Jin Z. Effects of bone marrow‑derived mesenchymal stem cell transplantation on chronic obstructive pulmonary disease/obstructive sleep apnea overlap syndrome in rats. Mol Med Rep 2019; 20:4665-4673. [PMID: 31702032 PMCID: PMC6797936 DOI: 10.3892/mmr.2019.10714] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 09/04/2019] [Indexed: 02/06/2023] Open
Abstract
Bone marrow-derived mesenchymal stem cells (BMSCs) possess potential therapeutic properties for treating patients with chronic obstructive pulmonary disease (COPD), which is characterized by emphysema and obstructive sleep apnea (OSA). However, their effects on overlap syndrome (OS) remain unclear. We investigated the potential therapeutic effects and possible mechanisms of BMSC transplantation in OS rats. To generate the OS model in rats, the animals underwent daily exposure to cigarette smoke and intermittent hypoxia. BMSCs were intravenously injected into rats. At 4 weeks post-transplantation, the severity of emphysema was assessed by lung hematoxylin and eosin (H&E) staining. The levels of oxidative stress and the malondialdehyde (MDA) and superoxide dismutase (SOD) contents in serum and lung were detected. The apoptosis of alveolar septal cells was also detected by TUNEL assay. Finally, we determined the expression of CD31 and VWF in lung tissues by an immunohistochemical (IHC) assay. It was found that BMSCs were able to migrate to the injured lung and aorta tissues. In lung tissues, transplanted BMSCs, some of which had differentiated into endotheliocytes, were found in the alveolar walls. The mean linear intercept (MLI) and pathological scores were higher and the mean alveolar number (MAN) was lower in the OS group than these parameters in the control group. These values were significantly reduced in the OS+BMSC group compared to those in the OS group. The MDA content was decreased and SOD activity was increased in the OS+BMSC group compared to those in the OS group. The apoptotic index of alveolar wall cells in the OS group was higher than that in the OS+BMSC group. The expression levels of CD31 and VWF in alveolar wall cells in the OS group were lower than those in the OS+BMSC group. These results indicate that BMSCs may inhibit the progression of emphysema in the OS model by differentiating into endotheliocytes and suppressing the apoptosis of endotheliocytes and oxidative stress. There is a possibility that the release of growth factors and structural support may be a determinant for the regenerative effects observed following treatment with BMSCs.
Collapse
Affiliation(s)
- Min Chen
- Department of Pneumology, The First People's Hospital‑Calmette Hospital of Kunming, Kunming, Yunnan 650224, P.R. China
| | - Zhaoming Huang
- Department of Pneumology, The First People's Hospital‑Calmette Hospital of Kunming, Kunming, Yunnan 650224, P.R. China
| | - Hong Bi
- Department of Pneumology, The First People's Hospital‑Calmette Hospital of Kunming, Kunming, Yunnan 650224, P.R. China
| | - Xinghua Pan
- Medical Innovation Research Center, 920 Hospital of PLA Joint Logistics Support Force, Kunming, Yunnan 650032, P.R. China
| | - Jian He
- Department of Pneumology, The First People's Hospital‑Calmette Hospital of Kunming, Kunming, Yunnan 650224, P.R. China
| | - Lewei He
- Department of Pneumology, The First People's Hospital‑Calmette Hospital of Kunming, Kunming, Yunnan 650224, P.R. China
| | - Xu He
- Department of Pneumology, The First People's Hospital‑Calmette Hospital of Kunming, Kunming, Yunnan 650224, P.R. China
| | - Junyi Du
- Department of Pneumology, The First People's Hospital‑Calmette Hospital of Kunming, Kunming, Yunnan 650224, P.R. China
| | - Kaihua Zhou
- Department of Pneumology, The First People's Hospital‑Calmette Hospital of Kunming, Kunming, Yunnan 650224, P.R. China
| | - Liyan Wang
- Department of Pneumology, The First People's Hospital‑Calmette Hospital of Kunming, Kunming, Yunnan 650224, P.R. China
| | - Qing Wang
- Department of Pneumology, The First People's Hospital‑Calmette Hospital of Kunming, Kunming, Yunnan 650224, P.R. China
| | - Xiang Guo
- Department of Pneumology, The First People's Hospital‑Calmette Hospital of Kunming, Kunming, Yunnan 650224, P.R. China
| | - Zhixian Jin
- Department of Pneumology, The First People's Hospital‑Calmette Hospital of Kunming, Kunming, Yunnan 650224, P.R. China
| |
Collapse
|
21
|
Adipose Tissue-Derived Stem Cells Have the Ability to Differentiate into Alveolar Epithelial Cells and Ameliorate Lung Injury Caused by Elastase-Induced Emphysema in Mice. Stem Cells Int 2019; 2019:5179172. [PMID: 31281377 PMCID: PMC6590553 DOI: 10.1155/2019/5179172] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 03/20/2019] [Accepted: 05/06/2019] [Indexed: 12/17/2022] Open
Abstract
Chronic obstructive pulmonary disease is a leading cause of mortality globally, with no effective therapy yet established. Adipose tissue-derived stem cells (ADSCs) are useful for ameliorating lung injury in animal models. However, whether ADSCs differentiate into functional cells remains uncertain, and no study has reported on the mechanism by which ADSCs improve lung functionality. Thus, in this study, we examined whether ADSCs differentiate into lung alveolar cells and are able to ameliorate lung injury caused by elastase-induced emphysema in model mice. Here, we induced ADSCs to differentiate into type 2 alveolar epithelial cells in vitro. We demonstrated that ADSCs can differentiate into type 2 alveolar epithelial cells in an elastase-induced emphysematous lung and that ADSCs improve pulmonary function of emphysema model mice, as determined with spirometry and 129Xe MRI. These data revealed a novel function for ADSCs in promoting repair of the damaged lung by direct differentiation into alveolar epithelial cells.
Collapse
|
22
|
Zhang X, Chen J, Xue M, Tang Y, Xu J, Liu L, Huang Y, Yang Y, Qiu H, Guo F. Overexpressing p130/E2F4 in mesenchymal stem cells facilitates the repair of injured alveolar epithelial cells in LPS-induced ARDS mice. Stem Cell Res Ther 2019; 10:74. [PMID: 30841904 PMCID: PMC6404316 DOI: 10.1186/s13287-019-1169-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 01/30/2019] [Accepted: 02/08/2019] [Indexed: 01/18/2023] Open
Abstract
Background Low differentiation rates of mesenchymal stem cells (MSCs) limit their therapeutic effects on patients in clinical studies. Our previous study demonstrated that overexpressing p130 or E2F4 affected the multipotential differentiation of MSCs, and the underlying mechanism was attributed to the regulation of the G1 phase. Improving the efficiency of MSC differentiation into epithelial cells is considered to be a new method. Therefore, this study was conducted to evaluate the effects of overexpressing p130 or E2F4 in MSCs on improving re-epithelization in lipopolysaccharide (LPS)-induced ARDS animals. Methods Mouse MSCs (mMSCs) stably transfected with p130 and E2F4 were transplanted intratracheally into LPS-induced ARDS mice. After 7 and 14 days, the mice were sacrificed, and the histopathology of the lungs was assessed by haematoxylin-eosin staining and lung injury scoring. Homing and differentiation of mMSCs were analysed by labelling and tracking mMSCs with NIR815 dye and immunofluorescent staining. Surfactant proteins A and C and occludin in the lungs were assessed by western blot. Permeability was evaluated by analysing the protein concentration of BALF using ELISA. Alveolar fluid clearance was assessed by absorbance measurements of BALF. Lung fibrosis was assessed by Masson’s trichrome staining and Ashcroft scoring. Results The engraftment of mMSCs overexpressing p130 or E2F4 led to attenuated histopathological impairment of the lung tissue, and the lung injury scores of the LPS+mBM-MSC-p130 and LPS+mBM-MSC-E2F4 groups were also decreased (p < 0.05). Overexpression of p130 or E2F4 also increased the retention of mMSCs in the lung (p < 0.05), increased differentiation into type II alveolar epithelial cells (p < 0.05), and improved alveolar epithelial permeability (p < 0.05). Additionally, mMSCs overexpressing p130 or E2F4 inhibited lung fibrosis according to the deposition of collagen and the fibrosis score in the lungs (p < 0.05). Conclusion Overexpressing p130 or E2F4 in mMSCs could further improve the injured structure and function of epithelial cells in the lungs of ARDS mice as a result of improved differentiation of mMSCs into epithelial cells. Electronic supplementary material The online version of this article (10.1186/s13287-019-1169-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiwen Zhang
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, No.87 Dingjiaqiao Road, Gulou District, Nanjing, 210009, People's Republic of China
| | - Jianxiao Chen
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, No.87 Dingjiaqiao Road, Gulou District, Nanjing, 210009, People's Republic of China
| | - Ming Xue
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, No.87 Dingjiaqiao Road, Gulou District, Nanjing, 210009, People's Republic of China
| | - Yuying Tang
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, No.87 Dingjiaqiao Road, Gulou District, Nanjing, 210009, People's Republic of China
| | - Jingyuan Xu
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, No.87 Dingjiaqiao Road, Gulou District, Nanjing, 210009, People's Republic of China
| | - Ling Liu
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, No.87 Dingjiaqiao Road, Gulou District, Nanjing, 210009, People's Republic of China
| | - Yingzi Huang
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, No.87 Dingjiaqiao Road, Gulou District, Nanjing, 210009, People's Republic of China
| | - Yi Yang
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, No.87 Dingjiaqiao Road, Gulou District, Nanjing, 210009, People's Republic of China
| | - Haibo Qiu
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, No.87 Dingjiaqiao Road, Gulou District, Nanjing, 210009, People's Republic of China
| | - Fengmei Guo
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, No.87 Dingjiaqiao Road, Gulou District, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
23
|
Evaluation of the Immunomodulatory Effects of All-Trans Retinoic Acid Solid Lipid Nanoparticles and Human Mesenchymal Stem Cells in an A549 Epithelial Cell Line Model. Pharm Res 2019; 36:50. [DOI: 10.1007/s11095-019-2583-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 01/31/2019] [Indexed: 12/13/2022]
|
24
|
MIST1, an Inductive Signal for Salivary Amylase in Mesenchymal Stem Cells. Int J Mol Sci 2019; 20:ijms20030767. [PMID: 30759717 PMCID: PMC6387180 DOI: 10.3390/ijms20030767] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 02/05/2019] [Accepted: 02/05/2019] [Indexed: 01/01/2023] Open
Abstract
Sjögren’s syndrome (SjS) is an autoimmune disease that destroys the salivary glands and results in severe dry mouth. Mesenchymal stem cell (MSC) transplantation has been recently proposed as a promising therapy for restoring cells in multiple degenerative diseases. We have recently utilized advanced proteomics biochemical assays to identify the key molecules involved in the mesenchymal-epithelial transition (MET) of co-cultured mouse bone-marrow-derived MSCs mMSCs with primary salivary gland cells. Among the multiple transcription factors (TFs) that were differentially expressed, two major TFs were selected: muscle, intestine, and stomach expression-1 (MIST1) and transcription factor E2a (TCF3). These factors were assessed in the current study for their ability to drive the expression of acinar cell marker, alpha-salivary amylase 1 (AMY1), and ductal cell marker, cytokeratin19 (CK19), in vitro. Overexpression of MIST1-induced AMY1 expression while it had little effect on CK19 expression. In contrast, TCF3 induced neither of those cellular markers. Furthermore, we have identified that mMSCs express muscarinic-type 3 receptor (M3R) mainly in the cytoplasm and aquaporin 5 (AQP5) in the nucleus. While MIST1 did not alter M3R levels in mMSCs, a TCF3 overexpression downregulated M3R expressions in mMSCs. The mechanisms for such differential regulation of glandular markers by these TFs warrant further investigation.
Collapse
|
25
|
CXCR4-Overexpressing Umbilical Cord Mesenchymal Stem Cells Enhance Protection against Radiation-Induced Lung Injury. Stem Cells Int 2019; 2019:2457082. [PMID: 30867667 PMCID: PMC6379846 DOI: 10.1155/2019/2457082] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/31/2018] [Accepted: 11/07/2018] [Indexed: 12/13/2022] Open
Abstract
Less quantity of transplanted mesenchymal stem cells (MSCs) influences the therapeutic effects on radiation-induced lung injury (RILI). Previous studies have demonstrated that MSCs overexpressing Chemokine (C-X-C motif) receptor 4 (CXCR4) could increase the quantity of transplanted cells to local tissues. In the present study, we conducted overexpressing CXCR4 human umbilical cord mesenchymal stem cell (HUMSC) therapy for RILI. C57BL mice received single dose of thoracic irradiation with 13 Gy of X-rays and then were administered saline, control HUMSCs, or CXCR4-overexpressing HUMSCs via tail vein. Transfection with CXCR4 enhanced the quantity of transplanted HUMSCs in the radiation-induced injured lung tissues. CXCR4-overexpressing HUMSCs not only improved histopathological changes but also decreased the radiation-induced expression of SDF-1, TGF-β1, α-SMA, and collagen I and inhibited the radiation-induced decreased expression of E-cadherin. Transplanted CXCR4-overexpressing HUMSCs also could express pro-SP-C, indicated adopting the feature of ATII. These finding suggests that CXCR4-overexpressing HUMSCs enhance the protection against RILI and may be a promising strategy for RILI treatment.
Collapse
|
26
|
Syndecan-2-positive, Bone Marrow-derived Human Mesenchymal Stromal Cells Attenuate Bacterial-induced Acute Lung Injury and Enhance Resolution of Ventilator-induced Lung Injury in Rats. Anesthesiology 2019; 129:502-516. [PMID: 29979191 DOI: 10.1097/aln.0000000000002327] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
WHAT WE ALREADY KNOW ABOUT THIS TOPIC WHAT THIS ARTICLE TELLS US THAT IS NEW: BACKGROUND:: Human mesenchymal stromal cells demonstrate promise for acute respiratory distress syndrome, but current studies use highly heterogenous cell populations. We hypothesized that a syndecan 2 (CD362)-expressing human mesenchymal stromal cell subpopulation would attenuate Escherichia coli-induced lung injury and enhance resolution after ventilator-induced lung injury. METHODS In vitro studies determined whether CD362 human mesenchymal stromal cells could modulate pulmonary epithelial inflammation, wound healing, and macrophage phagocytosis. Two in vivo rodent studies determined whether CD362 human mesenchymal stromal cells attenuated Escherichia coli-induced lung injury (n = 10/group) and enhanced resolution of ventilation-induced injury (n = 10/group). RESULTS CD362 human mesenchymal stromal cells attenuated cytokine-induced epithelial nuclear factor kappa B activation, increased epithelial wound closure, and increased macrophage phagocytosis in vitro. CD362 human mesenchymal stromal cells attenuated Escherichia coli-induced injury in rodents, improving arterial oxygenation (mean ± SD, 83 ± 9 vs. 60 ± 8 mmHg, P < 0.05), improving lung compliance (mean ± SD: 0.66 ± 0.08 vs. 0.53 ± 0.09 ml · cm H2O, P < 0.05), reducing bacterial load (median [interquartile range], 1,895 [100-3,300] vs. 8,195 [4,260-8,690] colony-forming units, P < 0.05), and decreasing structural injury compared with vehicle. CD362 human mesenchymal stromal cells were more effective than CD362 human mesenchymal stromal cells and comparable to heterogenous human mesenchymal stromal cells. CD362 human mesenchymal stromal cells enhanced resolution after ventilator-induced lung injury in rodents, restoring arterial oxygenation (mean ± SD: 113 ± 11 vs. 89 ± 11 mmHg, P < 0.05) and lung static compliance (mean ± SD: 0.74 ± 0.07 vs. 0.45 ± 0.07 ml · cm H2O, P < 0.05), resolving lung inflammation, and restoring histologic structure compared with vehicle. CD362 human mesenchymal stromal cells efficacy was at least comparable to heterogenous human mesenchymal stromal cells. CONCLUSIONS A CD362 human mesenchymal stromal cell population decreased Escherichia coli-induced pneumonia severity and enhanced recovery after ventilator-induced lung injury.
Collapse
|
27
|
Abstract
The lung is often overlooked as a metabolically active organ, yet biochemical studies have long demonstrated that glucose utilization surpasses that of many other organs, including the heart, kidney, and brain. For most cells in the lung, energy consumption is relegated to performing common cellular tasks, like mRNA transcription and protein translation. However, certain lung cell populations engage in more specialized types of energy-consuming behaviors, such as the beating of cilia or the production of surfactant. While many extrapulmonary diseases are now linked to abnormalities in cellular metabolism, the pulmonary community has only recently embraced the concept of metabolic dysfunction as a driver of respiratory pathology. Herein, we provide an overview of the major metabolic pathways in the lung and discuss how cells sense and adapt to low-energy states. Moreover, we review some of the emerging evidence that links alterations in cellular metabolism to the pathobiology of several common respiratory diseases.
Collapse
Affiliation(s)
- Gang Liu
- Division of Pulmonary, Allergy and Critical Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | - Ross Summer
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA;
| |
Collapse
|
28
|
Coppolino I, Ruggeri P, Nucera F, Cannavò MF, Adcock I, Girbino G, Caramori G. Role of Stem Cells in the Pathogenesis of Chronic Obstructive Pulmonary Disease and Pulmonary Emphysema. COPD 2018; 15:536-556. [DOI: 10.1080/15412555.2018.1536116] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Irene Coppolino
- Dipartimento di Scienze Biomediche, Unità Operativa Complessa di Pneumologia, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università degli Studi di Messina, Messina, Italy
| | - Paolo Ruggeri
- Dipartimento di Scienze Biomediche, Unità Operativa Complessa di Pneumologia, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università degli Studi di Messina, Messina, Italy
| | - Francesco Nucera
- Dipartimento di Scienze Biomediche, Unità Operativa Complessa di Pneumologia, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università degli Studi di Messina, Messina, Italy
| | - Mario Francesco Cannavò
- Dipartimento di Scienze Biomediche, Unità Operativa Complessa di Pneumologia, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università degli Studi di Messina, Messina, Italy
| | - Ian Adcock
- Airways Disease Section, National Heart and Lung Institute, Royal Brompton Hospital Biomedical Research Unit, Imperial College, London, UK
| | - Giuseppe Girbino
- Dipartimento di Scienze Biomediche, Unità Operativa Complessa di Pneumologia, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università degli Studi di Messina, Messina, Italy
| | - Gaetano Caramori
- Dipartimento di Scienze Biomediche, Unità Operativa Complessa di Pneumologia, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università degli Studi di Messina, Messina, Italy
| |
Collapse
|
29
|
Shao Y, Zhou F, He D, Zhang L, Shen J. Overexpression of CXCR7 promotes mesenchymal stem cells to repair phosgene-induced acute lung injury in rats. Biomed Pharmacother 2018; 109:1233-1239. [PMID: 30551373 DOI: 10.1016/j.biopha.2018.10.108] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 10/11/2018] [Accepted: 10/20/2018] [Indexed: 12/14/2022] Open
Abstract
Phosgene exposure may result in acute lung injury (ALI) with high mortality. Emerging evidence suggests that mesenchymal stem cells (MSCs) have a therapeutic potential against ALI. CXC chemokine receptor 7 (CXCR7) has been identified as a receptor of stromal-cell-derived factor 1 (SDF1) involved in MSC migration and may be an important mediator of the therapeutic effects of MSCs on ALI. In our study, we initially constructed a lentiviral vector overexpressing CXCR7 and then successfully transduced it into rat bone marrow-derived MSCs (resulting in MSCs-CXCR7). We found that ALI and the wet-to-dry ratio significantly decreased in the phosgene-exposed rats after administration of MSCs-CXCR7 or MSCs-GFP. Indeed, treatment with MSCs-CXCR7 caused further improvement. Moreover, injection of MSCs-CXCR7 significantly facilitated MSC homing to injured lung tissue. Meanwhile, overexpression of CXCR7 promoted differentiation of MSCs into type II alveolar epithelial (AT II) cells and enhanced the ability of MSCs to modulate the inflammatory response in phosgene-induced ALI. Taken together, our findings suggest that CXCR7-overexpressing MSCs may markedly facilitate treatment of phosgene-induced ALI (P-ALI) in rats.
Collapse
Affiliation(s)
- Yiru Shao
- Center of Emergency & Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai 201508, China; Medical Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai 201508, China; Medical Research Center for Chemical Injury, Emergency and Critical Care of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Fangqing Zhou
- Center of Emergency & Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai 201508, China; Medical Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai 201508, China; Medical Research Center for Chemical Injury, Emergency and Critical Care of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Daikun He
- Center of Emergency & Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai 201508, China; Medical Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai 201508, China; Medical Research Center for Chemical Injury, Emergency and Critical Care of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Lin Zhang
- Center of Emergency & Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai 201508, China; Medical Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai 201508, China; Medical Research Center for Chemical Injury, Emergency and Critical Care of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Jie Shen
- Center of Emergency & Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai 201508, China; Medical Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai 201508, China; Medical Research Center for Chemical Injury, Emergency and Critical Care of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai 201508, China.
| |
Collapse
|
30
|
Su X, Yang L, Yin Y, Huang J, Qiao F, Fang Y, Yu L, Wang Y, Zhou K, Wang J. Bone marrow mesenchymal stem cells tune the differentiation of myeloid-derived suppressor cells in bleomycin-induced lung injury. Stem Cell Res Ther 2018; 9:253. [PMID: 30257700 PMCID: PMC6158827 DOI: 10.1186/s13287-018-0983-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 08/13/2018] [Accepted: 08/15/2018] [Indexed: 01/28/2023] Open
Abstract
Background Bone marrow mesenchymal stem cells (BMSC) transfer has been attempted as a therapeutic strategy in experimental lung injury and fibrosis. Reduction of neutrophilic infiltration is one of the mechanisms involved in this effect. However, the mechanisms by which BMSC modulate neutrophil remains unknown. Methods and results Exposure of mice to bleomycin (BLM) resulted in significant accumulation of cells that express neutrophilic markers Gr-1HighCD11b+Ly-6GHighF4/80―CD115―CD49d―. These cells lacked immunosuppressive activity and could not be defined as myeloid-derived suppressor cells (MDSC). When BMSC were administrated to BLM-treated mice, they tuned the differentiation of Gr-1HighCD11b+ toward Gr-1LowCD11b+ cells. Gr-1LowCD11b+ cells exhibited unsegmented nuclei and expressed F4/80, Ly-6C, CD49d, and CD115 markers. These cells had potent immunosuppressive activity and thus could be defined as monocytic MDSC. As a result of such immunoregulation, BMSC mediated a decrease of pro-inflammatory products and amelioration of lung injury in BLM-treated mice. Further study using antibody array showed increased expression of macrophage colony-stimulating factor (M-CSF) in BMSC-treated mice. Accumulation of Gr-1LowCD11b+ cells in BMSC-treated mice was abrogated in M-CSF neutralizing mice. The beneficial effect of BMSC was independent of the ability of the cells to engraft in lung and in vitro coculture study of BMSC with Gr-1+CD11b+ cells showed that the induction of Gr-1LowCD11b+ cells by BMSC was independent of cell-cell contact. Conclusions These results document the generation of Gr-1HighCD11b+ cells in BLM-treated mice, and suggest that BMSC tune the differentiation of Gr-1HighCD11b+ toward Gr-1LowCD11b+ cells and therefore inhibit the progression of BLM-induced lung injury. Electronic supplementary material The online version of this article (10.1186/s13287-018-0983-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- XiaoSan Su
- Biomedical Research Center, Affiliated Calmette Hospital of Kunming Medical University, 504 Qing Nian Road, Kunming, Yunnan, 650011, People's Republic of China
| | - Liu Yang
- Biomedical Research Center, Affiliated Calmette Hospital of Kunming Medical University, 504 Qing Nian Road, Kunming, Yunnan, 650011, People's Republic of China
| | - YanFeng Yin
- Biomedical Research Center, Affiliated Calmette Hospital of Kunming Medical University, 504 Qing Nian Road, Kunming, Yunnan, 650011, People's Republic of China
| | - Jie Huang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, 295 Xi Chang Road, Kunming, Yunnan, 650032, People's Republic of China
| | - Fei Qiao
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, 295 Xi Chang Road, Kunming, Yunnan, 650032, People's Republic of China
| | - Yu Fang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, 295 Xi Chang Road, Kunming, Yunnan, 650032, People's Republic of China
| | - Lu Yu
- Department of Pathology, Affiliated Calmette Hospital of Kunming Medical University, 504 Qing Nian Road, Kunming, Yunnan, 650011, People's Republic of China
| | - YinYin Wang
- Biomedical Research Center, Affiliated Calmette Hospital of Kunming Medical University, 504 Qing Nian Road, Kunming, Yunnan, 650011, People's Republic of China
| | - KaiHua Zhou
- Department of Respiratory Diseases, Affiliated Calmette Hospital of Kunming Medical University, 504 Qing Nian Road, Kunming, Yunnan, 650011, People's Republic of China
| | - Jun Wang
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, 295 Xi Chang Road, Kunming, Yunnan, 650032, People's Republic of China.
| |
Collapse
|
31
|
Kang DH, Park YS, Lee DY. Senotherapy for attenuation of cellular senescence in aging and organ implantation. J IND ENG CHEM 2018. [DOI: 10.1016/j.jiec.2017.08.053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
32
|
Machado MN, Mazzoli-Rocha F, Casquilho NV, Maron-Gutierrez T, Ortenzi VH, Morales MM, Fortunato RS, Zin WA. Bone Marrow-Derived Mononuclear Cell Therapy in Papain-Induced Experimental Pulmonary Emphysema. Front Physiol 2018. [PMID: 29515461 PMCID: PMC5826278 DOI: 10.3389/fphys.2018.00121] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Murine papain-induced emphysema is a model that reproduces many of the features found in patients. Bone marrow-derived mononuclear cells (BMMC) have already been used to repair the alveolar epithelium in respiratory diseases, but not in the papain model. Thus, we hypothesized that BMMC could prevent the pathophysiological processes in papain-induced experimental emphysema. Female BALB/c mice received intratracheal instillation of 50 μL of saline (S groups) or papain (P groups, 10 IU/50 μl of saline) on days 1 and 7 of the experimental protocol. On the 14th day, 2 × 106 BMMC of male BALB/c mice (SC21 and PC21) or saline (SS21 and PS21) were injected by the jugular vein. Analyses were done on days 14 (S14 and P14) and 21 (SS21, PS21, SC21, and PC21) of the protocol. qPCR evaluated the presence of the Y chromosome in the lungs of BMMC recipient animals. Functional residual capacity (FRC), alveolar diameter, cellularity, elastic fiber content, concentrations of TNF-α, IL-1β, IL-6, MIP-2, KC, IFN-γ, apoptosis, mRNA expression of the dual oxidase (DUOX1 and DUOX2), production of H2O2 and DUOX activity were evaluated in lung tissue. We did not detect the Y chromosome in recipients' lungs. FRC, alveolar diameter, polymorphonuclear cells (PMN) and levels of KC, MIP-2, and IFN-γ increased in P14 and PS21 groups; the changes in the latter were reverted by BMMC. TNF-α, IL-1β e IL-6 were similar in all groups. The amount of elastic fibers was smaller in P14 and PS21 than in other groups, and BMMC did not increase it in PC21 mice. PS21 animals showed increased DUOX activity and mRNA expression for DUOX1 and 2. Cell therapy reverted the activity of DUOX and mRNA expression of DUOX1. BMMC reduced mRNA expression of DUOX2. Apoptosis index was elevated in PS21 mice, which was reduced by cell therapy in PC21. Static compliance, viscoelastic component of elastance and pressure to overcome viscoelasticity were increased in P14 and PS21 groups. These changes and the high resistive pressure found on day 21 were reverted by BMMC. In conclusion, BMMC showed potent anti-inflammatory, antiapoptotic, antioxidant, and restorative roles in papain-triggered pulmonary emphysema.
Collapse
Affiliation(s)
- Mariana N Machado
- Laboratory of Respiration Physiology, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Flavia Mazzoli-Rocha
- Laboratory of Respiration Physiology, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Natália V Casquilho
- Laboratory of Respiration Physiology, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Victor H Ortenzi
- Laboratory of Molecular Radiobiology, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo M Morales
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rodrigo S Fortunato
- Laboratory of Molecular Radiobiology, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Walter A Zin
- Laboratory of Respiration Physiology, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
33
|
Liu J, Wang W, Liu F, Li Z. Pediatric acute respiratory distress syndrome - current views. Exp Ther Med 2018; 15:1775-1780. [PMID: 29434764 PMCID: PMC5776650 DOI: 10.3892/etm.2017.5628] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 11/29/2017] [Indexed: 12/18/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) mainly involves acute respiratory failure. In addition to this affected patients feel progressive arterial hypoxemia, dyspnea, and a marked increase in the work of breathing. The only clinical solution for the above pathological state is ventilation. Mechanical ventilation is necessary to support life in ARDs but it itself worsen lung injury and the term is known clinically as ‘ventilation induced lung injury’ (VILI). At the cellular level, respiratory epithelial cells are subjected to cyclic stretch, i.e. repeated cycles of positive and negative strain, during normal tidal ventilation. In aerated areas of diseased lungs, or even normal lungs subjected to injurious positive pressure mechanical ventilation, the cells are at risk of being over distended, and worsening injury by disrupting the alveolar epithelial barrier. Further, hypercapnic acidosis (HCA) in itself confers protection from stretch injury, potentially via a mechanisms involving inhibition of nuclear factor κB (NF-κB), a transcription factor central to inflammation, injury and repair. Mesenchymal stem cells are the latest in the field and are being investigated as a possible therapy for ARDS.
Collapse
Affiliation(s)
- Jinfeng Liu
- Department of Neonatology, Xuzhou Chlidren's Hospital, Xuzhou, Jiangsu 221002, P.R. China
| | - Wei Wang
- Department of Neonatology, Xuzhou Chlidren's Hospital, Xuzhou, Jiangsu 221002, P.R. China
| | - Fengli Liu
- Department of Neonatology, Xuzhou Chlidren's Hospital, Xuzhou, Jiangsu 221002, P.R. China
| | - Zhenguang Li
- Department of Neonatology, Xuzhou Chlidren's Hospital, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
34
|
The promise of mesenchymal stem cell therapy for acute respiratory distress syndrome. J Trauma Acute Care Surg 2018; 84:183-191. [PMID: 29019797 DOI: 10.1097/ta.0000000000001713] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This review describes the current state of the science on mesenchymal stem cell (MSC) treatment for acute lung injury (ALI). The general characteristics, regenerative potential, and mechanism of action of MSCs are first presented. Next, particular emphasis is placed on the application of MSCs for the treatment of acute respiratory distress syndrome (ARDS) in preclinical and clinical studies. Finally, we discuss current challenges and future directions in the field presented from a clinician-researcher perspective. The objective of this work is to provide the readership with a current review of the literature discussing the hurdles and overall promise of MSCs as therapeutic interventions for the treatment of ARDS.
Collapse
|
35
|
Epithelial chimerism in lung tissue after allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant 2017; 53:474-477. [PMID: 29269810 DOI: 10.1038/s41409-017-0050-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 08/23/2017] [Accepted: 11/15/2017] [Indexed: 11/08/2022]
|
36
|
Papaccio F, Paino F, Regad T, Papaccio G, Desiderio V, Tirino V. Concise Review: Cancer Cells, Cancer Stem Cells, and Mesenchymal Stem Cells: Influence in Cancer Development. Stem Cells Transl Med 2017; 6:2115-2125. [PMID: 29072369 PMCID: PMC5702541 DOI: 10.1002/sctm.17-0138] [Citation(s) in RCA: 220] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Accepted: 10/04/2017] [Indexed: 12/21/2022] Open
Abstract
Tumors are composed of different types of cancer cells that contribute to tumor heterogeneity. Among these populations of cells, cancer stem cells (CSCs) play an important role in cancer initiation and progression. Like their stem cells counterpart, CSCs are also characterized by self-renewal and the capacity to differentiate. A particular population of CSCs is constituted by mesenchymal stem cells (MSCs) that differentiate into cells of mesodermal characteristics. Several studies have reported the potential pro-or anti-tumorigenic influence of MSCs on tumor initiation and progression. In fact, MSCs are recruited to the site of wound healing to repair damaged tissues, an event that is also associated with tumorigenesis. In other cases, resident or migrating MSCs can favor tumor angiogenesis and increase tumor aggressiveness. This interplay between MSCs and cancer cells is fundamental for cancerogenesis, progression, and metastasis. Therefore, an interesting topic is the relationship between cancer cells, CSCs, and MSCs, since contrasting reports about their respective influences have been reported. In this review, we discuss recent findings related to conflicting results on the influence of normal and CSCs in cancer development. The understanding of the role of MSCs in cancer is also important in cancer management. Stem Cells Translational Medicine 2017;6:2115-2125.
Collapse
Affiliation(s)
- Federica Papaccio
- Dipartimento Medico‐Chirurgico di Internistica Clinica e Sperimentale “F. Magrassi”, Università degli Studi della Campania ‘L. Vanvitelli’NaplesItaly
| | - Francesca Paino
- Dipartimento di Medicina Sperimentale, Sezione di Sezione di BiotecnologieIstologia Medica e Biologia Molecolare, Università degli Studi della Campania ‘L. Vanvitelli’NaplesItaly
| | - Tarik Regad
- The John van Geest Cancer Research Centre, School of Science and TechnologyNottingham Trent UniversityNottinghamUnited Kingdom
- Dipartimento di Biochimica, Biofisica, e Patologia GeneraleUniversità degli Studi della Campania ‘L. Vanvitelli’NaplesItaly
| | - Gianpaolo Papaccio
- Dipartimento di Medicina Sperimentale, Sezione di Sezione di BiotecnologieIstologia Medica e Biologia Molecolare, Università degli Studi della Campania ‘L. Vanvitelli’NaplesItaly
| | - Vincenzo Desiderio
- Dipartimento di Medicina Sperimentale, Sezione di Sezione di BiotecnologieIstologia Medica e Biologia Molecolare, Università degli Studi della Campania ‘L. Vanvitelli’NaplesItaly
| | - Virginia Tirino
- Dipartimento di Medicina Sperimentale, Sezione di Sezione di BiotecnologieIstologia Medica e Biologia Molecolare, Università degli Studi della Campania ‘L. Vanvitelli’NaplesItaly
| |
Collapse
|
37
|
Potential contribution of alveolar epithelial type I cells to pulmonary fibrosis. Biosci Rep 2017; 37:BSR20171301. [PMID: 29026006 PMCID: PMC5696455 DOI: 10.1042/bsr20171301] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 09/28/2017] [Accepted: 09/29/2017] [Indexed: 12/13/2022] Open
Abstract
Pulmonary fibrosis (PF) is characterized by inflammation and fibrosis of the interstitium and destruction of alveolar histoarchitecture ultimately leading to a fatal impairment of lung function. Different concepts describe either a dominant role of inflammatory pathways or a disturbed remodeling of resident cells of the lung parenchyma during fibrogenesis. Further, a combination of both the mechanisms has been postulated. The present review emphasizes the particular involvement of alveolar epithelial type I cells in all these processes, their contribution to innate immune/inflammatory functions and maintenance of proper alveolar barrier functions. Amongst the different inflammatory and repair events the purinergic receptor P2X7, an ATP-gated cationic channel that regulates not only apoptosis, necrosis, autophagy, and NLPR3 inflammosome activation, but also the turnover of diverse tight junction (TJ) and water channel proteins, seems to be essential for the stability of alveolar barrier integrity and for the interaction with protective factors during lung injury.
Collapse
|
38
|
Zhang S, Jiang W, Ma L, Liu Y, Zhang X, Wang S. Nrf2 transfection enhances the efficacy of human amniotic mesenchymal stem cells to repair lung injury induced by lipopolysaccharide. J Cell Biochem 2017; 119:1627-1636. [PMID: 28905450 DOI: 10.1002/jcb.26322] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 08/02/2017] [Indexed: 12/23/2022]
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are clinical emergencies with no effective pharmaceutical treatment. This study aims to determine the protective effects of Nrf2-transfected human amniotic mesenchymal stem cells (hAMSCs) against lipopolysaccharide (LPS)-induced lung injury in mice. hAMSCs stably transfected with Nrf2 or green fluorescent protein control were transplanted into male C57BL/6 mice via the tail vein 4 h after intratracheal instillation of LPS. At 3, 7, and 14 days after cell transplantation, total lung injury score (the Smith score) was determined by hematoxylin and eosin staining. Lung fibrosis was assessed by Masson's trichrome staining. Alveolar epithelial apoptosis was determined by terminal deoxynucleotidyl transferase dUTP nick end labeling staining. The plasma levels of interleukin (IL)-1β, IL-6, and IL-10 were determined by enzyme-linked immunosorbent assays (ELISA). The homing and differentiation of hAMSCs into type II alveolar epithelial (AT II) cells were examined by immunofluorescent staining and/or western blot analysis. Nrf2, mRNA, and protein expression in lungs were examined by qRT-PCR and western blot analysis, and DNA-binding activity of Nrf2 was detected by ELISA. We found that, compared with control hAMSCs, treatment with Nrf2-overexpressing hAMSCs led to further reduced lung injury, lung fibrosis, and inflammation in LPS-challenged mice. Nrf2-overexpressing hAMSCs also exhibited increased cell retention in the lung, more efficient differentiation into AT II cells, and more prominent effects on the increased mRNA and protein expression as well as DNA-binding activity of Nrf2 than control. These results support Nrf2-overexpressing hAMSCs as a potential cell-based therapy for clinical ALI/ARDS.
Collapse
Affiliation(s)
- Shouqin Zhang
- Department of Critical Care Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wei Jiang
- Department of Critical Care Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lijie Ma
- Department of Critical Care Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuhao Liu
- Department of Critical Care Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiangyu Zhang
- Department of Critical Care Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Sheng Wang
- Department of Critical Care Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
39
|
Ding Q, Liu G, Zeng Y, Zhu J, Liu Z, Jiang J, Huang J. Glycogen synthase kinase‑3β inhibitor reduces LPS‑induced acute lung injury in mice. Mol Med Rep 2017; 16:6715-6721. [PMID: 28901469 PMCID: PMC5865788 DOI: 10.3892/mmr.2017.7469] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 05/08/2017] [Indexed: 01/11/2023] Open
Abstract
The aim of the present study was to examine the role of Wnt signaling in lipopolysaccharide (LPS)‑induced acute respiratory distress syndrome (ARDS). ARDS was induced by LPS and compared in mice treated with either glycogen synthase kinase‑3β inhibitor (GSKI) or PBS. The protein expression levels of interleukin (IL)‑6, IL‑8, tumor necrosis factor (TNF)‑α, IL‑17, IL‑18 and IL‑1β in the bronchoalveolar lavage fluid (BALF) were examined using murine cytokine‑specific enzyme‑linked immunosorbent assays. The accumulation of neutrophils and macrophages in the BALF were detected using flow cytometry. The extent of pathological lesions was evaluated using an immunohistochemical assay. The differentiation of mesenchymal stem cells (MSCs) into type II alveolar (ATII) epithelial cells was analyzed using immunofluorescence staining. Treatment with GSKI led to maintained body weights and survival in mice with LPS‑induced ARDS. Treatment with GSKI effectively reduced the levels of total protein, albumin, IgM and keratinocyte growth factor in the BALF. Smith scores showed that GSKI significantly alleviated LPS‑induced lung injury. GSKI also functioned to reduce inflammatory cell accumulation and pro‑inflammatory cytokine secretion. Finally, it was found that GSKI promoted the differentiation of MSCs into ATII epithelial cells in vivo. Taken together, the GSKI‑treated mice exhibited reduced acute lung injury through inhibited intra‑fluid inflammatory cell infiltration and decreased expression of pro‑inflammatory cytokines, and GSKI increased the differentiation of MSCs into ATII epithelial cells.
Collapse
Affiliation(s)
- Qi Ding
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Jiangsu 215006, P.R. China
| | - Gaoqin Liu
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Yuanyuan Zeng
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Jiangsu 215006, P.R. China
| | - Jianjie Zhu
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Jiangsu 215006, P.R. China
| | - Zeyi Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Jiangsu 215006, P.R. China
| | - Junhong Jiang
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Jiangsu 215006, P.R. China
| | - Jianan Huang
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Jiangsu 215006, P.R. China
| |
Collapse
|
40
|
Beers MF, Moodley Y. When Is an Alveolar Type 2 Cell an Alveolar Type 2 Cell? A Conundrum for Lung Stem Cell Biology and Regenerative Medicine. Am J Respir Cell Mol Biol 2017; 57:18-27. [PMID: 28326803 DOI: 10.1165/rcmb.2016-0426ps] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Generating mature, differentiated, adult lung cells from pluripotent cells, such as induced pluripotent stem cells and embryonic stem cells, offers the hope of both generating disease-specific in vitro models and creating definitive and personalized therapies for a host of debilitating lung parenchymal and airway diseases. With the goal of advancing lung-regenerative medicine, several groups have developed and reported on protocols using defined media, coculture with mesenchymal components, or sequential treatments mimicking lung development, to obtain distal lung epithelial cells from stem cell precursors. However, there remains significant controversy about the degree of differentiation of these cells compared with their primary counterparts, coupled with a lack of consistency or uniformity in assessing the resultant phenotypes. Given the inevitable, exponential expansion of these approaches and the probable, but yet-to-emerge second and higher generation techniques to create such assets, we were prompted to pose the question, what makes a lung epithelial cell a lung epithelial cell? More specifically for this Perspective, we also posed the question, what are the minimum features that constitute an alveolar type (AT) 2 epithelial cell? In addressing this, we summarize a body of work spanning nearly five decades, amassed by a series of "lung epithelial cell biology pioneers," which carefully describes well characterized molecular, functional, and morphological features critical for discriminately assessing an AT2 phenotype. Armed with this, we propose a series of core criteria to assist the field in confirming that cells obtained following a differentiation protocol are indeed mature and functional AT2 epithelial cells.
Collapse
Affiliation(s)
- Michael F Beers
- 1 Lung Epithelial Biology Laboratories, Penn Center for Pulmonary Biology, Pulmonary and Critical Care Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Yuben Moodley
- 2 University of Western Australia, Harry Perkins Research Institute, Fiona Stanley Hospital, Murdoch, Western Australia, Australia
| |
Collapse
|
41
|
Zhu L, Fu X, Chen X, Han X, Dong P. M2 macrophages induce EMT through the TGF-β/Smad2 signaling pathway. Cell Biol Int 2017; 41:960-968. [PMID: 28493530 DOI: 10.1002/cbin.10788] [Citation(s) in RCA: 139] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 05/08/2017] [Indexed: 12/11/2022]
Abstract
IPF is characterized by fibroblast accumulation, collagen deposition, and ECM remodeling, with myofibroblasts believed to be the effector cell type. Myofibroblasts develop due to EMT of lung alveolar epithelial cells, which can be induced by TGF-β. M2 macrophages, a macrophage subpopulation, secrete large amounts of TGF-β. To clarify the relationship between IPF, EMT, TGF-β, and M2 macrophages, a bleomycin-induced pulmonary fibrosis mouse model was used. Seventeen days after mice were treated with bleomycin, the successful establishment of a pulmonary fibrosis model was confirmed by HE stain and Masson's trichrome stain. We found evidence in support of EMT, such as elevated protein levels of α-SMA in lung tissue and decreased levels of E-cadherin and CK-18. Additionally, increased TGF-β levels and TGF-β/Smad2 signaling activation was observed. Macrophages were recruited to pulmonary alveoli. Alveolar macrophages were phenotyped and identified as M2 macrophages, with up-regulated CD206 on the cell surfaces. For in vitro studies, we treated RAW 264.7 cells with IL-4 for 24 h, and the cells were then utilized as M2 macrophages. TGF-β levels increased significantly in the culture supernatant. Forty-eight hours after lung epithelial cells (MLE-12) were co-cultured with the M2 macrophages, the expression of α-SMA increased, and E-cadherin and CK-18 decreased. When a TGF-β receptor inhibitor, LY2109761 was used, the EMT induced by M2 macrophages was blocked. In conclusion, we demonstrated that M2 macrophages induce EMT through the TGF-β/Smad2 signaling pathway.
Collapse
Affiliation(s)
- Liangying Zhu
- Immunology and Reproductive Biology Laboratory, Medical School, Nanjing University, Hankou Road 22, Nanjing, 210093, China.,The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| | - Xiao Fu
- Immunology and Reproductive Biology Laboratory, Medical School, Nanjing University, Hankou Road 22, Nanjing, 210093, China.,The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| | - Xiang Chen
- Immunology and Reproductive Biology Laboratory, Medical School, Nanjing University, Hankou Road 22, Nanjing, 210093, China
| | - Xiaodong Han
- Immunology and Reproductive Biology Laboratory, Medical School, Nanjing University, Hankou Road 22, Nanjing, 210093, China
| | - Ping Dong
- Immunology and Reproductive Biology Laboratory, Medical School, Nanjing University, Hankou Road 22, Nanjing, 210093, China
| |
Collapse
|
42
|
Wei L, Zhang J, Yang ZL, You H. Extracellular superoxide dismutase increased the therapeutic potential of human mesenchymal stromal cells in radiation pulmonary fibrosis. Cytotherapy 2017; 19:586-602. [PMID: 28314668 DOI: 10.1016/j.jcyt.2017.02.359] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 01/31/2017] [Accepted: 02/16/2017] [Indexed: 01/22/2023]
Abstract
BACKGROUND AIMS Pulmonary fibrosis induced by irradiation is a significant problem of radiotherapy in cancer patients. Extracellular superoxide dismutase (SOD3) is found to be predominantly and highly expressed in the extracellular matrix of lung and plays a pivotal role against oxidative damage. Early administration of mesenchymal stromal cells (MSCs) has been demonstrated to reduce fibrosis of damaged lung. However, injection of MSCs at a later stage would be involved in fibrosis development. The present study aimed to determine whether injection of human umbilical cord-derived MSCs (UC-MSCs) over-expressing SOD3 at the established fibrosis stage would have beneficial effects in a mice model of radiation pulmonary fibrosis. METHODS Herein, pulmonary fibrosis in mice was induced using Cobalt-60 (60Co) irradiator with 20 Gy, followed by intravenous injection of UC-MSCs, transduced or not to express SOD3 at 2 h (early delivery) and 60 day (late delivery) post-irradiation, respectively. RESULTS Our results demonstrated that the early administration of UC-MSCs could attenuate the microscopic damage, reduce collagen deposition, inhibit (myo)fibroblast proliferation, reduce inflammatory cell infiltration, protect alveolar type II (AE2) cell injury, prevent oxidative stress and increase antioxidant status, and reduce pro-fibrotic cytokine level in serum. Furthermore, the early treatment with SOD3-infected UC-MSCs resulted in better improvement. However, we failed to observe the therapeutic effects of UC-MSCs, transduced to express SOD3, during established fibrosis. CONCLUSION Altogether, our results demonstrated that the early treatment with UC-MSCs alone significantly reduced radiation pulmonary fibrosis in mice through paracrine effects, with further improvement by administration of SOD3-infected UC-MSCs, suggesting that SOD3-infected UC-MSCs may be a potential cell-based gene therapy to treat clinical radiation pulmonary fibrosis.
Collapse
Affiliation(s)
- Li Wei
- Key Laboratory of Birth Defects and Reproductive Health of National Health and Family Planning Commission, Chongqing Population and Family Planning Science and Technology Research Institute, Chongqing, China
| | - Jing Zhang
- Oncology Department, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - Zai-Liang Yang
- Department of Breast and Thyroid, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China.
| | - Hua You
- Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China.
| |
Collapse
|
43
|
Li X, Wang Y, An G, Liang D, Zhu Z, Lian X, Niu P, Guo C, Tian L. Bone marrow mesenchymal stem cells attenuate silica-induced pulmonary fibrosis via paracrine mechanisms. Toxicol Lett 2017; 270:96-107. [DOI: 10.1016/j.toxlet.2017.02.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 02/16/2017] [Accepted: 02/18/2017] [Indexed: 12/21/2022]
|
44
|
Zhu H, Xiong Y, Xia Y, Zhang R, Tian D, Wang T, Dai J, Wang L, Yao H, Jiang H, Yang K, Liu E, Shi Y, Fu Z, Gao L, Zou L. Therapeutic Effects of Human Umbilical Cord-Derived Mesenchymal Stem Cells in Acute Lung Injury Mice. Sci Rep 2017; 7:39889. [PMID: 28051154 PMCID: PMC5209685 DOI: 10.1038/srep39889] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 11/29/2016] [Indexed: 02/05/2023] Open
Abstract
The incidence and mortality of acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) are still very high, but stem cells show some promise for its treatment. Here we found that intratracheal administration of human umbilical cord-mesenchymal stem cells (UC-MSCs) significantly improved survival and attenuated the lung inflammation in lipopolysaccharide (LPS)-induced ALI mice. We also used the proteins-chip and bioinformatics to analyze interactions between UC-MSCs treatment and immune-response alternations of ALI mice. Then we demonstrated that UC-MSCs could inhibit the inflammatory response of mouse macrophage in ALI mice, as well as enhance its IL-10 expression. We provide data to support the concept that the therapeutic capacity of UC-MSCs for ALI was primarily through paracrine secretion, particularly of prostaglandin-E2 (PGE2). Furthermore, we showed that UC-MSCs might secrete a panel of factors including GM-CSF, IL-6 and IL-13 to ameliorate ALI. Our study suggested that UC-MSCs could protect LPS-induced ALI model by immune regulation and paracrine factors, indicating that UC-MSCs should be a promising strategy for ALI/ARDS.
Collapse
Affiliation(s)
- Hua Zhu
- Pediatrics Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
- Department of Pediatrics, First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Yi Xiong
- Pediatrics Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Yunqiu Xia
- Pediatrics Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
- Department of Respiratory Medicine, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Rong Zhang
- Pediatrics Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Daiyin Tian
- Pediatrics Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
- Department of Respiratory Medicine, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Ting Wang
- Pediatrics Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
- Department of Respiratory Medicine, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Jihong Dai
- Pediatrics Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
- Department of Respiratory Medicine, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Lijia Wang
- Pediatrics Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Hongbing Yao
- Pediatrics Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
- Department of Otorhinolaryngology, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Hong Jiang
- Department of Pediatrics, First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Ke Yang
- Pediatrics Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing 400014, China
| | - Enmei Liu
- Pediatrics Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
- Department of Respiratory Medicine, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Yujun Shi
- Laboratoryof Pathology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhou Fu
- Pediatrics Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
- Department of Respiratory Medicine, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing 400014, China
| | - Li Gao
- Pediatrics Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
- Department of Otorhinolaryngology, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing 400014, China
| | - Lin Zou
- Pediatrics Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
- Chongqing Engineering Research Center of Stem Cell Therapy, Chongqing 400014, China
- Center for Clinical Molecular Medicine, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
| |
Collapse
|
45
|
Maria OM, Maria AM, Ybarra N, Jeyaseelan K, Lee S, Perez J, Shalaby MY, Lehnert S, Faria S, Serban M, Seuntjens J, El Naqa I. Mesenchymal Stem Cells Adopt Lung Cell Phenotype in Normal and Radiation-induced Lung Injury Conditions. Appl Immunohistochem Mol Morphol 2016. [PMID: 26200842 DOI: 10.1097/pai.0000000000000180] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Lung tissue exposure to ionizing irradiation can invariably occur during the treatment of a variety of cancers leading to increased risk of radiation-induced lung disease (RILD). Mesenchymal stem cells (MSCs) possess the potential to differentiate into epithelial cells. However, cell culture methods of primary type II pneumocytes are slow and cannot provide a sufficient number of cells to regenerate damaged lungs. Moreover, effects of ablative radiation doses on the ability of MSCs to differentiate in vitro into lung cells have not been investigated yet. Therefore, an in vitro coculture system was used, where MSCs were physically separated from dissociated lung tissue obtained from either healthy or high ablative doses of 16 or 20 Gy whole thorax irradiated rats. Around 10±5% and 20±3% of cocultured MSCs demonstrated a change into lung-specific Clara and type II pneumocyte cells when MSCs were cocultured with healthy lung tissue. Interestingly, in cocultures with irradiated lung biopsies, the percentage of MSCs changed into Clara and type II pneumocytes cells increased to 40±7% and 50±6% at 16 Gy irradiation dose and 30±5% and 40±8% at 20 Gy irradiation dose, respectively. These data suggest that MSCs to lung cell differentiation is possible without cell fusion. In addition, 16 and 20 Gy whole thorax irradiation doses that can cause varying levels of RILD, induced different percentages of MSCs to adopt lung cell phenotype compared with healthy lung tissue, providing encouraging outlook for RILD therapeutic intervention for ablative radiotherapy prescriptions.
Collapse
Affiliation(s)
- Ola M Maria
- *Medical Physics Unit, Department of Oncology, Radiation Oncology Division, McGill University, Montreal General Hospital ‡Department of Oncology, Radiation Oncology Division, McGill University Health Centre ∥International Baccalaureate, Marymount Academy, Montreal, QC, Canada †Department of Oral Biology, Faculty of Dentistry, Mansoura University, Mansoura §Faculty of Medicine, Tanta University, Tanta, Egypt
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Hu Y, Xiong LL, Zhang P, Wang TH. Microarray expression profiles of genes in lung tissues of rats subjected to focal cerebral ischemia-induced lung injury following bone marrow-derived mesenchymal stem cell transplantation. Int J Mol Med 2016; 39:57-70. [PMID: 27922691 PMCID: PMC5179184 DOI: 10.3892/ijmm.2016.2819] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 11/21/2016] [Indexed: 02/05/2023] Open
Abstract
Ischemia-induced stroke is the most common disease of the nervous system and is associated with a high mortality rate worldwide. Cerebral ischemia may lead to remote organ dysfunction, particular in the lungs, resulting in lung injury. Nowadays, bone marrow-derived mesenchymal stem cells (BMSCs) are widely studied in clinical trials as they may provide an effective solution to the treatment of neurological and cardiac diseases; however, the underlying molecular mechanisms remain unknown. In this study, a model of permanent focal cerebral ischemia-induced lung injury was successfully established and confirmed by neurological evaluation and lung injury scores. We demonstrated that the transplantation of BMSCs (passage 3) via the tail vein into the lung tissues attenuated lung injury. In order to elucidate the underlying molecular mechanisms, we analyzed the gene expression profiles in lung tissues from the rats with focal cerebral ischemia and transplanted with BMSCs using a Gene microarray. Moreover, the Gene Ontology database was employed to determine gene function. We found that the phosphoinositide 3-kinase (PI3K)-AKT signaling pathway, transforming growth factor-β (TGF-β) and platelet-derived growth factor (PDGF) were downregulated in the BMSC transplantation groups, compared with the control group. These results suggested that BMSC transplantation may attenuate lung injury following focal cerebral ischemia and that this effect is associated with the downregulation of TGF-β, PDGF and the PI3K-AKT pathway.
Collapse
Affiliation(s)
- Yue Hu
- Department of Anesthesiology, Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Liu-Lin Xiong
- Department of Anesthesiology, Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Piao Zhang
- Institute of Neuroscience, Kunming Medical University, Kunming, Yunnan 650031, P.R. China
| | - Ting-Hua Wang
- Department of Anesthesiology, Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
47
|
Perrini C, Strillacci MG, Bagnato A, Esposti P, Marini MG, Corradetti B, Bizzaro D, Idda A, Ledda S, Capra E, Pizzi F, Lange-Consiglio A, Cremonesi F. Microvesicles secreted from equine amniotic-derived cells and their potential role in reducing inflammation in endometrial cells in an in-vitro model. Stem Cell Res Ther 2016; 7:169. [PMID: 27863532 PMCID: PMC5114748 DOI: 10.1186/s13287-016-0429-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 09/22/2016] [Accepted: 10/21/2016] [Indexed: 12/24/2022] Open
Abstract
Background It is known that a paracrine mechanism exists between mesenchymal stem cells and target cells. This process may involve microvesicles (MVs) as an integral component of cell-to-cell communication. Methods In this context, this study aims to understand the efficacy of MVs in in-vitro endometrial stressed cells in view of potential healing in in-vivo studies. For this purpose, the presence and type of MVs secreted by amniotic mesenchymal stem cells (AMCs) were investigated and the response of endometrial cells to MVs was studied using a dose-response curve at different concentrations and times. Moreover, the ability of MVs to counteract the in vitro stress in endometrial cells induced by lipopolysaccharide was studied by measuring the rate of apoptosis and cell proliferation, the expression of some pro-inflammatory genes such as tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), interleukin 1β (IL-1β), and metalloproteinases (MMP) 1 and 13, and the release of some pro- or anti-inflammatory cytokines. Results MVs secreted by the AMCs ranged in size from 100 to 200 nm. The incorporation of MVs was gradual over time and peaked at 72 h. MVs reduced the apoptosis rate, increased cell proliferation values, downregulated pro-inflammatory gene expression, and decreased the secretion of pro-inflammatory cytokines. Conclusion Our data suggest that some microRNAs could contribute to counteracting in-vivo inflammation of endometrial tissue.
Collapse
Affiliation(s)
- Claudia Perrini
- Large Animal Hospital, Reproduction Unit, Università degli Studi di Milano, Via dell'Università 6, 26900, Lodi, Italy
| | | | - Alessandro Bagnato
- Department of Veterinary Medicine, Università degli Studi di Milano, Milano, Italy
| | - Paola Esposti
- Large Animal Hospital, Reproduction Unit, Università degli Studi di Milano, Via dell'Università 6, 26900, Lodi, Italy
| | - Maria Giovanna Marini
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Bruna Corradetti
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Davide Bizzaro
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Antonella Idda
- Department of Veterinary Medicine, Università degli Studi di Sassari, Sassari, Italy
| | - Sergio Ledda
- Department of Veterinary Medicine, Università degli Studi di Sassari, Sassari, Italy
| | - Emanuele Capra
- Institute of Biology and Agricultural Biotechnology-CNR, Milano, Italy
| | - Flavia Pizzi
- Institute of Biology and Agricultural Biotechnology-CNR, Milano, Italy
| | - Anna Lange-Consiglio
- Large Animal Hospital, Reproduction Unit, Università degli Studi di Milano, Via dell'Università 6, 26900, Lodi, Italy.
| | - Fausto Cremonesi
- Large Animal Hospital, Reproduction Unit, Università degli Studi di Milano, Via dell'Università 6, 26900, Lodi, Italy.,Department of Veterinary Medicine, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
48
|
Doster DL, Jensen AR, Khaneki S, Markel TA. Mesenchymal stromal cell therapy for the treatment of intestinal ischemia: Defining the optimal cell isolate for maximum therapeutic benefit. Cytotherapy 2016; 18:1457-1470. [PMID: 27745788 DOI: 10.1016/j.jcyt.2016.08.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 07/16/2016] [Accepted: 08/02/2016] [Indexed: 02/06/2023]
Abstract
Intestinal ischemia is a devastating intraabdominal emergency that often necessitates surgical intervention. Mortality rates can be high, and patients who survive often have significant long-term morbidity. The implementation of traditional medical therapies to prevent or treat intestinal ischemia have been sparse over the last decade, and therefore, the use of novel therapies are becoming more prevalent. Cellular therapy using mesenchymal stromal cells is one such treatment modality that is attracting noteworthy attention in the scientific community. Several groups have seen benefit with cellular therapy, but the optimal cell line has not been identified. The purpose of this review is to: 1) Review the mechanism of intestinal ischemia and reperfusion injury, 2) Identify the mechanisms of how cellular therapy may be therapeutic for this disease, and 3) Compare various MSC tissue sources to maximize potential therapeutic efficacy in the treatment of intestinal I/R diseases.
Collapse
Affiliation(s)
- Dominique L Doster
- Department of Surgery, Indiana University Health, Indianapolis, IN, USA; The Indiana University School of Medicine, Indianapolis, IN, USA
| | - Amanda R Jensen
- Department of Surgery, Indiana University Health, Indianapolis, IN, USA; The Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sina Khaneki
- Department of Surgery, Indiana University Health, Indianapolis, IN, USA; The Indiana University School of Medicine, Indianapolis, IN, USA
| | - Troy A Markel
- Department of Surgery, Indiana University Health, Indianapolis, IN, USA; The Indiana University School of Medicine, Indianapolis, IN, USA; Section of Pediatric Surgery, Indiana University Health, Indianapolis, IN, USA; Riley Hospital for Children, Indiana University Health, Indianapolis, IN, USA.
| |
Collapse
|
49
|
Skrahin A, Jenkins HE, Hurevich H, Solodovnikova V, Isaikina Y, Klimuk D, Rohava Z, Skrahina A. Effectiveness of a novel cellular therapy to treat multidrug-resistant tuberculosis. J Clin Tuberc Other Mycobact Dis 2016; 4:21-27. [PMID: 27284577 PMCID: PMC4894747 DOI: 10.1016/j.jctube.2016.05.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Introduction We urgently need novel treatments for multidrug-resistant tuberculosis (MDR-TB). Autologous mesenchymal stromal cell (MSC) infusion is one such possibility due to its potential to repair damaged lung tissue and boost immune responses. We aimed to assess the effectiveness of MSC to improve outcomes among MDR-TB patients. Methods We analyzed outcomes for 108 Belarussian MDR-TB patients receiving chemotherapy. Thirty-six patients (“cases”) also had MSCs extracted, cultured and re-infused (average time from chemotherapy start to infusion was 49 days); another 36 patients were “study controls”. We identified another control group: 36 patients from the Belarussian surveillance database (“surveillance controls”) 1:1 matched to cases. Results Of the cases, 81% had successful outcomes versus 42% of surveillance controls and 39% of study controls. Successful outcome odds were 6.5 (95% Confidence Interval: 1.2–36.2, p = 0.032) times greater for cases than surveillance controls (age-adjusted). Radiological improvement was more likely in cases than study controls. Culture analysis prior to infusion demonstrated a poorer initial prognosis in cases, yet despite this they had better outcomes than the control groups. Conclusion MSC treatment could vastly improve outcomes for MDR-TB patients. Our findings could revolutionize therapy options and have strong implications for future directions of MDR-TB therapy research.
Collapse
Affiliation(s)
- Aliaksandr Skrahin
- Clinical Department, Republican Research and Practical Centre for Pulmonology and TB, Minsk, Belarus
- Department of Intensive Care and Anesthesiology, Belarus State Medical University, Minsk, Belarus
| | - Helen E. Jenkins
- Division of Global Health Equity, Brigham and Women's Hospital, Boston, USA
- Harvard Medical School, Boston, USA
- Corresponding author at: Department of Biostatistics, Boston University School of Public Health, 801 Massachusetts Avenue, Boston, MA 02118, USA
| | - Henadz Hurevich
- Clinical Department, Republican Research and Practical Centre for Pulmonology and TB, Minsk, Belarus
| | - Varvara Solodovnikova
- Clinical Department, Republican Research and Practical Centre for Pulmonology and TB, Minsk, Belarus
| | - Yanina Isaikina
- Laboratory of Cellular Biotechnology and Cytotherapy, Belarussian Research Centre for Paediatric Oncology, Haematology and Immunology, Minsk, Belarus
| | - Dzmitri Klimuk
- Department of Monitoring and Evaluation, Republican Research and Practical Centre for Pulmonology and TB, Minsk, Belarus
| | - Zoya Rohava
- Laboratory Department, Republican Research and Practical Centre for Pulmonology and TB, Minsk, Belarus
| | - Alena Skrahina
- Clinical Department, Republican Research and Practical Centre for Pulmonology and TB, Minsk, Belarus
| |
Collapse
|
50
|
Li J, Huang S, Zhang J, Feng C, Gao D, Yao B, Wu X, Fu X. Mesenchymal stem cells ameliorate inflammatory cytokine-induced impairment of AT-II cells through a keratinocyte growth factor-dependent PI3K/Akt/mTOR signaling pathway. Mol Med Rep 2016; 13:3755-62. [PMID: 27035760 PMCID: PMC4838139 DOI: 10.3892/mmr.2016.5004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 02/04/2016] [Indexed: 01/18/2023] Open
Abstract
Lung epithelium restoration subsequent to injury is of concern in association with the outcomes of diverse inflammatory lung diseases. Previous studies have demonstrated that mesenchymal stem cells (MSCs) may promote epithelial repair subsequent to inflammatory injury, however the mechanism that mediates this effect remains unclear. The current study examined the role of MSCs in alveolar type II epithelial cell (AT-II cell) restoration subsequent to an inflammatory insult. AT-II cells were firstly exposed to inflammatory cytokines including tumor necrosis factor-α, interleukin (IL)-6 and IL-1β, then were co-cultured with MSCs in Transwell for 72 h. Cell proliferation, expression of surfactant protein A (SP-A) and expression of the α1 subunit were evaluated respectively by the Cell Counting Kit-8 assay, western blotting and semiquantitative reverse transcription-polymerase chain reaction. Keratinocyte growth factor (KGF) small interfering RNA (siRNA) was applied to knockdown the main cytoprotective factors in the MSCs. Subsequent to an inflammatory insult, AT-II cells were observed to be impaired, exhibiting the characteristics of injured cell morphology, reduced cell proliferation and reduced expression of SP-A and the α1 subunit. Co-culture with MSCs significantly ameliorated these cell impairments, while these benefits were weakened by the application of KGF siRNA. Simultaneously, expression levels of phosphorylated (p-) protein kinase B (AKT) and p-mammalian target of rapamycin (mTOR) in AT-II cells were upregulated by MSCs, suggesting activation of the phosphoinositide 3-kinase (PI3K) pathway. These data demonstrate that administration of MSCs to the inflammation-insulted AT-II cells may ameliorate the impairments through a KGF-dependent PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Jiwei Li
- Department of Thoracic and Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Sha Huang
- Key Laboratory of Wound Repair and Regeneration of People's Liberation Army, The First Affiliated Hospital, Trauma Center of Postgraduate Medical College, Beijing 100048, P.R. China
| | - Junhua Zhang
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Changjiang Feng
- Department of Thoracic and Cardiovascular Surgery, Peking University People's Hospital, Peking University, Beijing 100044, P.R. China
| | - Dongyun Gao
- Department of Oncology, Dongtai People's Hospital, Dongtai, Jiangsu 224200, P.R. China
| | - Bin Yao
- Key Laboratory of Wound Repair and Regeneration of People's Liberation Army, The First Affiliated Hospital, Trauma Center of Postgraduate Medical College, Beijing 100048, P.R. China
| | - Xu Wu
- Department of Thoracic and Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Xiaobing Fu
- Key Laboratory of Wound Repair and Regeneration of People's Liberation Army, The First Affiliated Hospital, Trauma Center of Postgraduate Medical College, Beijing 100048, P.R. China
| |
Collapse
|