51
|
Suvakov S, Richards C, Nikolic V, Simic T, McGrath K, Krasnodembskaya A, McClements L. Emerging Therapeutic Potential of Mesenchymal Stem/Stromal Cells in Preeclampsia. Curr Hypertens Rep 2020; 22:37. [PMID: 32291521 DOI: 10.1007/s11906-020-1034-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Preeclampsia is a dangerous pregnancy condition affecting both the mother and offspring. It is a multifactorial disease with poorly understood pathogenesis, lacking effective treatments. Maternal immune response, inflammation and oxidative stress leading to endothelial dysfunction are the most prominent pathogenic processes implicated in preeclampsia development. Here, we give a detailed overview of the therapeutic applications and mechanisms of mesenchymal stem/stromal cells (MSCs) as a potential new treatment for preeclampsia. RECENT FINDINGS MSCs have gained growing attention due to low immunogenicity, easy cultivation and expansion in vitro. Accumulating evidence now suggests that MSCs act primarily through their secretomes facilitating paracrine signalling that leads to potent immunomodulatory, pro-angiogenic and regenerative therapeutic effects. MSCs have been studied in different animal models of preeclampsia demonstrating promising result, which support further investigations into the therapeutic effects and mechanisms of MSC-based therapies in preeclampsia, steering these therapies into clinical trials.
Collapse
Affiliation(s)
- S Suvakov
- Department of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA.,Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - C Richards
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | - V Nikolic
- Department of Pharmacology and Toxicology, Medical Faculty, University of Nis, Nis, Serbia
| | - T Simic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, Belgrade, Serbia.,Serbian Academy of Sciences and Arts, Belgrade, Serbia
| | - K McGrath
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | - A Krasnodembskaya
- The Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - L McClements
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia.
| |
Collapse
|
52
|
Behnke J, Kremer S, Shahzad T, Chao CM, Böttcher-Friebertshäuser E, Morty RE, Bellusci S, Ehrhardt H. MSC Based Therapies-New Perspectives for the Injured Lung. J Clin Med 2020; 9:jcm9030682. [PMID: 32138309 PMCID: PMC7141210 DOI: 10.3390/jcm9030682] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 02/25/2020] [Accepted: 02/28/2020] [Indexed: 12/11/2022] Open
Abstract
Chronic lung diseases pose a tremendous global burden. At least one in four people suffer from severe pulmonary sequelae over the course of a lifetime. Despite substantial improvements in therapeutic interventions, persistent alleviation of clinical symptoms cannot be offered to most patients affected to date. Despite broad discrepancies in origins and pathomechanisms, the important disease entities all have in common the pulmonary inflammatory response which is central to lung injury and structural abnormalities. Mesenchymal stem cells (MSC) attract particular attention due to their broadly acting anti-inflammatory and regenerative properties. Plenty of preclinical studies provided congruent and convincing evidence that MSC have the therapeutic potential to alleviate lung injuries across ages. These include the disease entities bronchopulmonary dysplasia, asthma and the different forms of acute lung injury and chronic pulmonary diseases in adulthood. While clinical trials are so far restricted to pioneering trials on safety and feasibility, preclinical results point out possibilities to boost the therapeutic efficacy of MSC application and to take advantage of the MSC secretome. The presented review summarizes the most recent advances and highlights joint mechanisms of MSC action across disease entities which provide the basis to timely tackle this global disease burden.
Collapse
Affiliation(s)
- Judith Behnke
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Feulgenstrasse 12, 35392 Gießen, Germany; (J.B.); (S.K.); (T.S.); (C.-M.C.)
| | - Sarah Kremer
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Feulgenstrasse 12, 35392 Gießen, Germany; (J.B.); (S.K.); (T.S.); (C.-M.C.)
| | - Tayyab Shahzad
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Feulgenstrasse 12, 35392 Gießen, Germany; (J.B.); (S.K.); (T.S.); (C.-M.C.)
| | - Cho-Ming Chao
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Feulgenstrasse 12, 35392 Gießen, Germany; (J.B.); (S.K.); (T.S.); (C.-M.C.)
- Department of Internal Medicine II, Universities of Giessen and Marburg Lung Center (UGMLC), Cardiopulmonary Institute (CPI), German Center for Lung Research (DZL), Aulweg 130, 35392 Giessen, Germany;
| | | | - Rory E. Morty
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, German Center for Lung Research (DZL), Ludwigstrasse 43, 61231 Bad Nauheim, Germany;
| | - Saverio Bellusci
- Department of Internal Medicine II, Universities of Giessen and Marburg Lung Center (UGMLC), Cardiopulmonary Institute (CPI), German Center for Lung Research (DZL), Aulweg 130, 35392 Giessen, Germany;
| | - Harald Ehrhardt
- Department of General Pediatrics and Neonatology, Justus-Liebig-University, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Feulgenstrasse 12, 35392 Gießen, Germany; (J.B.); (S.K.); (T.S.); (C.-M.C.)
- Correspondence: ; Tel.: +49-985-43400; Fax: +49-985-43419
| |
Collapse
|
53
|
Schmiedl A, Bokel K, Huhn V, Ionescu L, Zscheppang K, Dammann CEL. Bone marrow stem cells accelerate lung maturation and prevent the LPS-induced delay of morphological and functional fetal lung development in the presence of ErbB4. Cell Tissue Res 2020; 380:547-564. [PMID: 32055958 DOI: 10.1007/s00441-019-03145-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 11/18/2019] [Indexed: 12/28/2022]
Abstract
ErbB4 is a regulator in lung development and disease. Prenatal infection is an important risk factor for the delay of morphologic lung development, while promoting the maturation of the surfactant system. Bone marrow-derived mesenchymal stem cells (BMSCs) have the potential to prevent lung injury. We hypothesized that BMSCs in comparison with hematopoietic control stem cells (HPSCs) minimize the lipopolysaccharide (LPS)-induced lung injury only when functional ErbB4 receptor is present. We injected LPS and/or murine green fluorescent protein-labeled BMSCs or HPSCs into the amniotic cavity of transgenic ErbB4heart mothers at gestational day 17. Fetal lungs were analyzed 24 h later. BMSCs minimized significantly LPS-induced delay in morphological lung maturation consisting of a stereologically measured increase in mesenchyme and septal thickness and a decrease of future airspace and septal surface. This effect was more prominent and significant in the ErbB4heart+/- lungs, suggesting that the presence of functioning ErbB4 signaling is required. BMSC also diminished the LPS induced increase in surfactant protein (Sftp)a mRNA and decrease in Sftpc mRNA is only seen if ErbB4 is present. The reduction of morphological delay of lung development and of levels of immune-modulating Sftp was more pronounced in the presence of the ErbB4 receptor. Thus, ErbB4 may be required for the protective signaling of BMSCs.
Collapse
Affiliation(s)
- Andreas Schmiedl
- Institute of Functional and Applied Anatomy, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany.
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center of Lung Research (DZL), Hannover, Germany.
| | - Kyra Bokel
- Institute of Functional and Applied Anatomy, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Verena Huhn
- Department of Pediatric Pulmonology and Neonatology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Lavinia Ionescu
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Katja Zscheppang
- Department of Pediatric Pulmonology and Neonatology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Christiane E L Dammann
- Department of Pediatric Pulmonology and Neonatology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
- Division of Newborn Medicine, Department of Pediatrics, Floating Hospital for Children at Tufts Medical Center, Boston, MA, USA
- Graduate School for Biomedical Sciences, Tufts University, Boston, MA, USA
| |
Collapse
|
54
|
Purity Determines the Effect of Extracellular Vesicles Derived from Mesenchymal Stromal Cells. Cells 2020; 9:cells9020422. [PMID: 32059497 PMCID: PMC7072280 DOI: 10.3390/cells9020422] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/05/2020] [Accepted: 02/10/2020] [Indexed: 12/15/2022] Open
Abstract
Extracellular vesicles (EVs) have been recently identified as vital components of cell-based therapies based on the observation that conditioned media from cultured stromal cells reproduce some of the beneficial effects of intact cells. In order to obtain clinically active EVs derived from Mesenchymal Stromal Cells (MSCs) different procedures have been reported in the literature. Usually, non-confluent cells are incubated with culture medium for 48 h either with EV-depleted Fetal Bovine Serum (FBS) or without FBS. Our aim was to compare the effects of EVs isolated by ultracentrifugation from human umbilical cord MSC conditioned media obtained using these two conditions: with EV-depleted FBS (UC) or without FBS (UCw/o) on the mRNA expression levels of extracellular matrix related genes using the mouse chondrogenic cell line ATDC-5. We observed a deleterious effect on chondrogenic cells treated with UCw/o, showing higher mRNA expression levels of different metalloproteinases and decorin (Dcn) and lower collagen (Col1a1 and Col2a1) and aggrecan (Acan) mRNA levels. To elucidate whether this deleterious effect was induced by the EVs or by any proteins co-purified in the EV pellet, we used size exclusion chromatography (SEC) to further purify the EV pellet, obtaining an EV enriched fraction (EV or EVw/o) and a protein enriched fraction (Prot or Protw/o). Our results pointed that the negative effect on the chondrogenic cell line was due to the contaminant proteins coisolated with the EVs by ultracentrifugation and not from the EVs themselves. Thus, these results highlight the importance of working with well purified EV preparations to specifically achieve their therapeutic effect.
Collapse
|
55
|
Jafarinia M, Alsahebfosoul F, Salehi H, Eskandari N, Ganjalikhani-Hakemi M. Mesenchymal Stem Cell-Derived Extracellular Vesicles: A Novel Cell-Free Therapy. Immunol Invest 2020; 49:758-780. [PMID: 32009478 DOI: 10.1080/08820139.2020.1712416] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In recent years, mesenchymal stem/stromal cells (MSCs) have widely been considered as therapeutic tools in basic researches and clinical trials. Accumulating evidence supports the idea that MSCs perform their therapeutic roles in paracrine manner especially through trophic factors and extracellular vesicles (EVs). Compared to cells, EVs have several advantages to be used as therapeutic agents, such as they lack self-replicating capabilities, dangers of ectopic differentiation, and tumor formation, genetic instability, and cellular rejection by the immune system. Since the MSC-derived EVs (MSC-EVs) appear to exert similar therapeutic effects of their parent cells, such as ability to arrive themselves to the site of injury and immunomodulatory properties, MSC-EVs have been widely studied in many animal models, including kidney, liver, cardiovascular, immunological, and neurological diseases. Regarding this, MSC-EVs look to be a novel and interesting approach to be studied in clinical trials of different inflammatory diseases. In this review, we summarize the properties and applications of MSC-EVs in different diseases.
Collapse
Affiliation(s)
- Morteza Jafarinia
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences , Isfahan, Iran
| | - Fereshteh Alsahebfosoul
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences , Isfahan, Iran
| | - Hossein Salehi
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences , Isfahan, Iran
| | - Nahid Eskandari
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences , Isfahan, Iran
| | | |
Collapse
|
56
|
Gülhan PY, Ekici MS, Niyaz M, Gülhan M, Erçin ME, Ekici A, Aksoy N. Therapeutic Treatment with Abdominal Adipose Mesenchymal Cells Does Not Prevent Elastase-Induced Emphysema in Rats. Turk Thorac J 2020; 21:14-20. [PMID: 32163359 PMCID: PMC7020897 DOI: 10.5152/turkthoracj.2019.180136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 02/06/2019] [Indexed: 11/22/2022]
Abstract
OBJECTIVES Emphysema and chronic bronchitis have different pathophysiologies but both are significant components of chronic obstructive lung disease (COPD). The levels of Matrix metalloproteinase (MMP)-9 in the bronchoalveloar lavage fluid (BALF) and in serum indicate the presence of emphysema. Intratracheal administration of elastase has been used to create a rat model of emphysema. Adipose tissue-derived mesenchymal stem cells (MSC) have been postulated to prevent or reverse emphysema, however, this has not been examined in the rat model of elastase-induced emphysema. MATERIALS AND METHODS In this study, 31 Wistar albino rats aged 6-8 weeks and weighing 250-300 g were assessed. On day 1, the animals were treated intratracheally with 0.5 mL saline (control group, n=10), i.e., 0.5 mL saline solution containing 0.1 IU porcine pancreatic elastase (PPE) (Elastase group, n=12) or PPE plus MSC (Elastase-MSC group, n=9) was adminstered per animal. MSCs suspended in serum were injected via the caudal vein on day 21. At least 106 cells were injected. All animals were sacrificed on day 42 and the emphysema index (EI) was calculated, along with measuring the BALF and serum MMP-9 concentrations. RESULTS Porcine pancreatic elastase induced a significant degree of emphysema in the PPE groups as compared to the control group, which was determined by the EI index (p=0.008). This was not reversed by MSC treatment. The EI remained significantly low in comprison with the controls (p=0.001) and measured no different from the Elastase-treated animals. There was no statistically significant difference between the BALF and serum MMP-9 levels between the control and treatment groups. CONCLUSION Our findings suggest that therapeutic treatment with adipose tissue-derived MSC in rats has no effect on emphysema or on MMP9 expression, which is a known marker of emphysema.
Collapse
Affiliation(s)
- Pınar Yıldız Gülhan
- Department of Chest Diseases, Düzce University School of Medicine, Düzce, Turkey
| | - Mehmet Savaş Ekici
- Department of Chest Diseases, Kırıkkale University School of Medicine, Kırıkkale, Turkey
| | - Mehmet Niyaz
- Clinic of Cardiovascular Surgery, Bartın State Hospital, Bartın, Turkey
| | - Muhammet Gülhan
- Clinic of Infectious Diseases and Clinical Microbiology, Tosya State Hospital, Kastamonu, Turkey
| | - Mustafa Emre Erçin
- Department of Pathology, Karadeniz Technical University School of Medicine, Trabzon, Turkey
| | - Aydanur Ekici
- Department of Chest Diseases, Kırıkkale University School of Medicine, Kırıkkale, Turkey
| | - Nurkan Aksoy
- Clinic of Biochemistry, Yenimahalle State Hospital, Ankara, Turkey
| |
Collapse
|
57
|
Abstract
As the prevalence and impact of lung diseases continue to increase worldwide, new therapeutic strategies are desperately needed. Advances in lung-regenerative medicine, a broad field encompassing stem cells, cell-based therapies, and a range of bioengineering approaches, offer new insights into and new techniques for studying lung physiology and pathophysiology. This provides a platform for the development of novel therapeutic approaches. Applicability to chronic obstructive pulmonary disease of recent advances and applications in cell-based therapies, predominantly those with mesenchymal stromal cell-based approaches, and bioengineering approaches for lung diseases are reviewed.
Collapse
|
58
|
Li L, Wang R, Jia Y, Rong R, Xu M, Zhu T. Exosomes Derived From Mesenchymal Stem Cells Ameliorate Renal Ischemic-Reperfusion Injury Through Inhibiting Inflammation and Cell Apoptosis. Front Med (Lausanne) 2019; 6:269. [PMID: 31867333 PMCID: PMC6907421 DOI: 10.3389/fmed.2019.00269] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Accepted: 11/01/2019] [Indexed: 01/15/2023] Open
Abstract
This study aimed to investigate the underlying mechanism of mesenchymal stem cells (MSCs) on protection of renal ischemia reperfusion injury (IRI). Exosomes originated from MSCs (MSC-ex) were extracted according to the instructions of Total Exosome Isolation Reagent. Rats were divided into five groups: sham-operated, IRI, MSC, MSC-ex, and MSC-ex + RNAase group. MSCs or MSC-ex were injected via carotid artery. The renal function test and pathological detection were applied to determine the renoprotection of MSC-ex on IRI. Western blotting and quantitative reverse transcription polymerase chain reaction (RT-qPCR) were conducted to examine the levels of apoptosis-related proteins and inflammatory cytokines. Our results revealed that MSC-derived exosomes attenuated renal dysfunction, histologic damage, and decreased apoptosis. The expression levels of inflammatory cytokines, such as interleukin 6 (IL-6), tumor necrosis factor-α (TNF-α), nuclear factor kappa B (NF-κB), and interferon gamma (IFN-γ), were decreased by the MSC-ex treatment. The expression levels of caspase-9, cleaved caspase-3, Bax, and Bcl-2 caused by IR were also inhibited by MSC-ex. MSC-ex + RNAase group shared the similar pattern of changes with IRI group, likely due to the ability of RNA hydrolase to eliminate the function of exosomes. Our results demonstrated that exosomes originating from MSCs have protective effects on IRI via inhibiting cell apoptosis and inflammatory responses. Out findings may provide a new insight into therapeutic mechanism of MSCs on renal IRI.
Collapse
Affiliation(s)
- Long Li
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Rulin Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yichen Jia
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Ruiming Rong
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Ming Xu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Tongyu Zhu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| |
Collapse
|
59
|
Abstract
Introduction: Mesenchymal stem/stromal cells (MSCs) have been shown to improve lung function and survival in chronic inflammatory lung diseases, including asthma, chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), pulmonary arterial hypertension (PAH), and silicosis.Areas covered: This review covers rationale for the use of MSC therapy, along with preclinical studies and clinical trials with MSC therapy in chronic lung diseases.Expert opinion: MSC therapy holds promise for the treatment of chronic lung diseases, mainly when administered at early stages. In clinical trials, MSC administration was safe, but associated with limited effects on clinical outcomes. Further studies are required to elucidate unresolved issues, including optimal MSC source and dose, route of administration, and frequency (single vs. multiple-dose regimens). A better understanding of the mechanisms of MSC action, local microenvironment of each disease, and development of strategies to potentiate the beneficial effects of MSCs may improve outcomes.
Collapse
|
60
|
Lan YW, Yang JC, Yen CC, Huang TT, Chen YC, Chen HL, Chong KY, Chen CM. Predifferentiated amniotic fluid mesenchymal stem cells enhance lung alveolar epithelium regeneration and reverse elastase-induced pulmonary emphysema. Stem Cell Res Ther 2019; 10:163. [PMID: 31196196 PMCID: PMC6567664 DOI: 10.1186/s13287-019-1282-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 04/23/2019] [Accepted: 05/27/2019] [Indexed: 03/08/2023] Open
Abstract
INTRODUCTION Pulmonary emphysema is a major component of chronic obstructive pulmonary disease (COPD). Emphysema progression attributed not only to alveolar structure loss and pulmonary regeneration impairment, but also to excessive inflammatory response, proteolytic and anti-proteolytic activity imbalance, lung epithelial cells apoptosis, and abnormal lung remodeling. To ameliorate lung damage with higher efficiency in lung tissue engineering and cell therapy, pre-differentiating graft cells into more restricted cell types before transplantation could enhance their ability to anatomically and functionally integrate into damaged lung. In this study, we aimed to evaluate the regenerative and repair ability of lung alveolar epithelium in emphysema model by using lung epithelial progenitors which pre-differentiated from amniotic fluid mesenchymal stem cells (AFMSCs). METHODS Pre-differentiation of eGFP-expressing AFMSCs to lung epithelial progenitor-like cells (LEPLCs) was established under a modified small airway growth media (mSAGM) for 7-day induction. Pre-differentiated AFMSCs were intratracheally injected into porcine pancreatic elastase (PPE)-induced emphysema mice at day 14, and then inflammatory-, fibrotic-, and emphysema-related indices and pathological changes were assessed at 6 weeks after PPE administration. RESULTS An optimal LEPLCs pre-differentiation condition has been achieved, which resulted in a yield of approximately 20% lung epithelial progenitors-like cells from AFMSCs in a 7-day period. In PPE-induced emphysema mice, transplantation of LEPLCs significantly improved regeneration of lung tissues through integrating into the lung alveolar structure, relieved airway inflammation, increased expression of growth factors such as vascular endothelial growth factor (VEGF), and reduced matrix metalloproteinases and lung remodeling factors when compared with mice injected with AFMSCs. Histopathologic examination observed a significant amelioration in DNA damage in alveolar cells, detected by terminal deoxynucleotidyltransferase-mediated dUTP nick end labeling (TUNEL), the mean linear intercept, and the collagen deposition in the LEPLC-transplanted groups. CONCLUSION Transplantation of predifferentiated AFMSCs through intratracheal injection showed better alveolar regeneration and reverse elastase-induced pulmonary emphysema in PPE-induced pulmonary emphysema mice.
Collapse
Affiliation(s)
- Ying-Wei Lan
- Department of Life Sciences, College of Life Sciences, National Chung Hsing University, No. 250, Kuo Kuang Rd., Taichung, 402 Taiwan
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, 333 Taiwan
- Graduate Institute of Biomedical Sciences, Division of Biotechnology, College of Medicine, Chang Gung University, Taoyuan, 333 Taiwan
| | - Jing-Chan Yang
- Department of Life Sciences, College of Life Sciences, National Chung Hsing University, No. 250, Kuo Kuang Rd., Taichung, 402 Taiwan
| | - Chih-Ching Yen
- Department of Life Sciences, College of Life Sciences, National Chung Hsing University, No. 250, Kuo Kuang Rd., Taichung, 402 Taiwan
- Department of Internal Medicine, China Medical University Hospital, Taichung, 404 Taiwan
- College of Health Care, China Medical University, Taichung, 404 Taiwan
| | - Tsung-Teng Huang
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, 333 Taiwan
| | - Ying-Cheng Chen
- Department of Life Sciences, College of Life Sciences, National Chung Hsing University, No. 250, Kuo Kuang Rd., Taichung, 402 Taiwan
| | - Hsiao-Ling Chen
- Department of Bioresource, Da-Yeh University, Changhwa, 515 Taiwan
| | - Kowit-Yu Chong
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, 333 Taiwan
- Graduate Institute of Biomedical Sciences, Division of Biotechnology, College of Medicine, Chang Gung University, Taoyuan, 333 Taiwan
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan, 333 Taiwan
- Centre for Stem Cell Research, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, 43000 Kajang, Selangor Malaysia
| | - Chuan-Mu Chen
- Department of Life Sciences, College of Life Sciences, National Chung Hsing University, No. 250, Kuo Kuang Rd., Taichung, 402 Taiwan
- The iEGG and Animal Biotechnology Center, and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, 402 Taiwan
| |
Collapse
|
61
|
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: 11] [Impact Index Per Article: 2.2] [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
|
62
|
Skolasinski SD, Panoskaltsis-Mortari A. Lung tissue bioengineering for chronic obstructive pulmonary disease: overcoming the need for lung transplantation from human donors. Expert Rev Respir Med 2019; 13:665-678. [PMID: 31164014 DOI: 10.1080/17476348.2019.1624163] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Introduction: Chronic obstructive pulmonary disease (COPD) affects more than 380 million people, causing more than 3 million deaths annually worldwide. Despite this enormous burden, currently available therapies are largely limited to symptom control. Lung transplant is considered for end-stage disease but is severely limited by the availability of human organs. Furthermore, the pre-transplant course is a complex orchestration of locating and harvesting suitable lungs, and the post-transplant course is complicated by rejection and infection. Lung tissue bioengineering has the potential to relieve the organ shortage and improve the post-transplant course by generating patient-specific lungs for transplant. Additionally, emerging progenitor cell therapies may facilitate in vivo regeneration of pulmonary tissue, obviating the need for transplant. Areas Covered: We review several lung tissue bioengineering approaches including the recellularization of decellularized scaffolds, 3D bioprinting, genetically-engineered xenotransplantation, blastocyst complementation, and direct therapy with progenitor cells. Articles were identified by searching relevant terms (see Key Words) in the PubMed database and selected for inclusion based on novelty and uniqueness of their approach. Expert Opinion: Lung tissue bioengineering research is in the early stages. Of the methods reviewed, only direct cell therapy has been investigated in humans. We anticipate a minimum of 5-10 years before human therapy will be feasible.
Collapse
Affiliation(s)
- Steven D Skolasinski
- a Division of Pulmonary, Allergy, Critical Care and Sleep Medicine , University of Minnesota , Minneapolis , MN , USA
| | | |
Collapse
|
63
|
Neuhaus M, Munder A, Schipke J, Schmiedl A. Lung infection caused by Pseudomonas aeruginosa in a CD26/DPP4 deficient F344 rat model. Inflamm Res 2019; 68:529-544. [PMID: 31089745 PMCID: PMC7079754 DOI: 10.1007/s00011-019-01236-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 03/27/2019] [Accepted: 04/16/2019] [Indexed: 01/21/2023] Open
Abstract
Background Pseudomonas aeruginosa (PA) is the most important opportunistic pathogen in causing nosocomial infections and, furthermore, poses a permanent threat for severe chronic infections in patients with cystic fibrosis or COPD. The transmembrane protein CD26 with dipeptidyl peptidase-4 (DPP4) activity shows an increased expression in inflamed tissue. We tested whether CD26/DPP4 deficiency leads to reduced inflammation and decreased structural damage when infected with PA. Methods CD26/DPP4+ and CD26/DPP4− rats were instilled intratracheally with NaCl (controls) or with PA. Six hours later, bacterial distribution was detected with the in vivo imaging system 200 (IVIS). Lungs were then processed for molecular biology, light and electron microscopy and analyzed qualitatively, quantitatively and stereologically. Bacterial numbers were determined in homogenized lungs. Results Compared to saline treated controls, in both infected groups (1) the acinar airspace was significantly increased, (2) the volume density of the alveolar epithelium was significantly decreased, (3) the septal thickness was significantly reduced, (4) more than 40% of the alveolar epithelial surface was damaged, and up to 36% of the epithelial surface was covered with edema. In infected CD26− rats, the increase in lung weight was significantly less pronounced, the portion of edematous alveolar airspace was significantly lower and the part of edema interspersed with PA was decreased significantly. Conclusions CD26/DPP4 deficiency resulted in reduced pulmonary edema under sublethal PA infection, implicating a role for CD26 in infection progression. The partly pronounced structural damage may mask further possible influences of CD26 on the inflammatory response.
Collapse
Affiliation(s)
- M Neuhaus
- Institute of Functional and Applied Anatomy, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany.,Department of Oral and Maxillofacial Surgery, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - A Munder
- Clinic of Pediatric Pneumology, Allergology and Neonatology Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover Medical School, Hannover, Germany
| | - J Schipke
- Institute of Functional and Applied Anatomy, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover Medical School, Hannover, Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - A Schmiedl
- Institute of Functional and Applied Anatomy, Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany. .,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover Medical School, Hannover, Germany. .,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
64
|
Mesenchymal Stem Cell-Based Therapy of Inflammatory Lung Diseases: Current Understanding and Future Perspectives. Stem Cells Int 2019; 2019:4236973. [PMID: 31191672 PMCID: PMC6525794 DOI: 10.1155/2019/4236973] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 02/06/2019] [Accepted: 02/14/2019] [Indexed: 12/16/2022] Open
Abstract
During acute or chronic lung injury, inappropriate immune response and/or aberrant repair process causes irreversible damage in lung tissue and most usually results in the development of fibrosis followed by decline in lung function. Inhaled corticosteroids and other anti-inflammatory drugs are very effective in patients with inflammatory lung disorders, but their long-term use is associated with severe side effects. Accordingly, new therapeutic agents that will attenuate ongoing inflammation and, at the same time, promote regeneration of injured alveolar epithelial cells are urgently needed. Mesenchymal stem cells (MSCs) are able to modulate proliferation, activation, and effector function of all immune cells that play an important role in the pathogenesis of acute and chronic inflammatory lung diseases. In addition to the suppression of lung-infiltrated immune cells, MSCs have potential to differentiate into alveolar epithelial cells in vitro and, accordingly, represent new players in cell-based therapy of inflammatory lung disorders. In this review article, we described molecular mechanisms involved in MSC-based therapy of acute and chronic pulmonary diseases and emphasized current knowledge and future perspectives related to the therapeutic application of MSCs in patients suffering from acute respiratory distress syndrome, pneumonia, asthma, chronic obstructive pulmonary diseases, and idiopathic pulmonary fibrosis.
Collapse
|
65
|
Bernardini C, Bertocchi M, Zannoni A, Salaroli R, Tubon I, Dothel G, Fernandez M, Bacci ML, Calzà L, Forni M. Constitutive and LPS-stimulated secretome of porcine Vascular Wall-Mesenchymal Stem Cells exerts effects on in vitro endothelial angiogenesis. BMC Vet Res 2019; 15:123. [PMID: 31029157 PMCID: PMC6487069 DOI: 10.1186/s12917-019-1873-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 04/16/2019] [Indexed: 12/20/2022] Open
Abstract
Background MSCs secretome is under investigation as an alternative to whole-cell-based therapies, since it is enriched of bioactive molecules: growth factors, cytokines and chemokines. Taking into account the translational value of the pig model, the leading aim of the present paper was to characterize the secretome of porcine Vascular Wall–Mesenchymal Stem Cells (pVW-MSCs) and its change in presence of LPS stimulation. Moreover, considering the importance of angiogenesis in regenerative mechanisms, we analysed the effect of pVW-MSCs secretome on in vitro angiogenesis. Results Our results demonstrated that conditioned medium from unstimulated pVW-MSCs contained high levels of IL-8, GM-CSF, IFN-γ and other immunomodulatory proteins: IL-6 IL-18 IL-4 IL-2 IL-10. LPS modulates pVW-MSCs gene expression and secretome composition, in particular a significant increase of IL-6 and IL-8 was observed; conversely, the amount of GM-CSF, IFN-γ, IL-2, IL-4, IL-10 and IL-18 showed a significant transient decrease with the LPS stimulation. Conditioned medium from unstimulated pVW-MSCs induced in vitro endothelial angiogenesis, which is more evident when the conditioned medium was from LPS stimulated pVW-MSCs. Conclusions The lines of evidence here presented shed a light on possible future application of secretome derived by pVW-MSCs on research studies in translational regenerative medicine.
Collapse
Affiliation(s)
- Chiara Bernardini
- Department of Veterinary Medical Sciences DIMEVET, University of Bologna, Via Tolara di Sopra 50, 40064 Ozzano Emilia, Bologna, Italy.
| | - Martina Bertocchi
- Department of Veterinary Medical Sciences DIMEVET, University of Bologna, Via Tolara di Sopra 50, 40064 Ozzano Emilia, Bologna, Italy
| | - Augusta Zannoni
- Department of Veterinary Medical Sciences DIMEVET, University of Bologna, Via Tolara di Sopra 50, 40064 Ozzano Emilia, Bologna, Italy
| | - Roberta Salaroli
- Department of Veterinary Medical Sciences DIMEVET, University of Bologna, Via Tolara di Sopra 50, 40064 Ozzano Emilia, Bologna, Italy
| | - Irvin Tubon
- Department of Veterinary Medical Sciences DIMEVET, University of Bologna, Via Tolara di Sopra 50, 40064 Ozzano Emilia, Bologna, Italy.,Escuela de Enfermeria, Facultad de Ciencias Medicas, Universidad Regional Autónoma de Los Andes UNIANDES, Ambato, EC180150, Ecuador
| | - Giovanni Dothel
- Department of Medical and Surgical Sciences - DIMEC, University of Bologna, Bologna, Italy
| | - Mercedes Fernandez
- Department of Veterinary Medical Sciences DIMEVET, University of Bologna, Via Tolara di Sopra 50, 40064 Ozzano Emilia, Bologna, Italy
| | - Maria Laura Bacci
- Department of Veterinary Medical Sciences DIMEVET, University of Bologna, Via Tolara di Sopra 50, 40064 Ozzano Emilia, Bologna, Italy
| | - Laura Calzà
- Department of Pharmacy and Biotechnology - FaBiT, University of Bologna, Bologna, Italy
| | - Monica Forni
- Department of Veterinary Medical Sciences DIMEVET, University of Bologna, Via Tolara di Sopra 50, 40064 Ozzano Emilia, Bologna, Italy
| |
Collapse
|
66
|
Clinical Application of Stem/Stromal Cells in COPD. STEM CELL-BASED THERAPY FOR LUNG DISEASE 2019. [PMCID: PMC7121219 DOI: 10.1007/978-3-030-29403-8_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive life-threatening disease that is significantly increasing in prevalence and is predicted to become the third leading cause of death worldwide by 2030. At present, there are no true curative treatments that can stop the progression of the disease, and new therapeutic strategies are desperately needed. Advances in cell-based therapies provide a platform for the development of new therapeutic approaches in severe lung diseases such as COPD. At present, a lot of focus is on mesenchymal stem (stromal) cell (MSC)-based therapies, mainly due to their immunomodulatory properties. Despite increasing number of preclinical studies demonstrating that systemic MSC administration can prevent or treat experimental COPD and emphysema, clinical studies have not been able to reproduce the preclinical results and to date no efficacy or significantly improved lung function or quality of life has been observed in COPD patients. Importantly, the completed appropriately conducted clinical trials uniformly demonstrate that MSC treatment in COPD patients is well tolerated and no toxicities have been observed. All clinical trials performed so far, have been phase I/II studies, underpowered for the detection of potential efficacy. There are several challenges ahead for this field such as standardized isolation and culture procedures to obtain a cell product with high quality and reproducibility, administration strategies, improvement of methods to measure outcomes, and development of potency assays. Moreover, COPD is a complex pathology with a diverse spectrum of clinical phenotypes, and therefore it is essential to develop methods to select the subpopulation of patients that is most likely to potentially respond to MSC administration. In this chapter, we will discuss the current state of the art of MSC-based cell therapy for COPD and the hurdles that need to be overcome.
Collapse
|
67
|
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: 1.0] [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
|
68
|
Park JS, Kim HK, Kang EY, Cho R, Oh YM. Potential Therapeutic Strategy in Chronic Obstructive Pulmonary Disease Using Pioglitazone-Augmented Wharton's Jelly-Derived Mesenchymal Stem Cells. Tuberc Respir Dis (Seoul) 2018; 82:158-165. [PMID: 30302955 PMCID: PMC6435932 DOI: 10.4046/trd.2018.0044] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 06/20/2018] [Accepted: 07/17/2018] [Indexed: 02/06/2023] Open
Abstract
Background A recent study reported that mesenchymal stem cells possess potential cellular therapeutic properties for treating patients with chronic obstructive pulmonary disease, which is characterized by emphysema. We examined the potential therapeutic effect of Wharton's Jelly-derived mesenchymal stem cells (WJMSCs), following pretreatment with pioglitazone, in lung regeneration mouse emphysema models. Methods We used two mouse emphysema models, an elastase-induced model and a cigarette smoke-induced model. We intravenously injected WJMSCs (1×104/mouse) to mice, pretreated or not, with pioglitazone for 7 days. We measured the emphysema severity by mean linear intercepts (MLI) analysis using lung histology. Results Pioglitazone pretreated WJMSCs (pioWJMSCs) were associated with greater lung regeneration than non-augmented WJMSCs in the two mouse emphysema models. In the elastase-induced emphysema model, the MLIs were 59.02±2.42 µm (n=6), 72.80±2.87 µm (n=6), for pioWJMSCs injected mice, and non-augmented WJMSCs injected mice, respectively (p<0.01). Both pioWJMSCs and non-augmented WJMSCs showed regenerative effects in the cigarette smoke emphysema model (MLIs were 41.25±0.98 [n=6] for WJMSCs and38.97±0.61 µm [n=6] for pioWJMSCs) compared to smoking control mice (51.65±1.36 µm, n=6). The mean improvement of MLI appeared numerically better in pioWJMSCs than in non-augmented WJMSCs injected mice, but the difference did not reach the level of statistical significance (p=0.071). Conclusion PioWJMSCs may produce greater lung regeneration, compared to non-augmented WJMSCs, in a mouse emphysema model.
Collapse
Affiliation(s)
| | - Hyun Kuk Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Inje University Haeundae Paik Hospital, Busan, Korea
| | | | | | - Yeon Mok Oh
- Asan Institute for Life Sciences, Seoul, Korea.,Department of Pulmonary and Critical Care Medicine, University of Ulsan College of Medicine, Seoul, Korea.,Department of Pulmonary and Critical Care Medicine, Asan Medical Center, Seoul, Korea.
| |
Collapse
|
69
|
Rühle A, Huber PE, Saffrich R, Lopez Perez R, Nicolay NH. The current understanding of mesenchymal stem cells as potential attenuators of chemotherapy-induced toxicity. Int J Cancer 2018; 143:2628-2639. [PMID: 29931767 DOI: 10.1002/ijc.31619] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/18/2018] [Accepted: 05/22/2018] [Indexed: 12/18/2022]
Abstract
Chemotherapeutic agents are part of the standard treatment algorithms for many malignancies; however, their application and dosage are limited by their toxic effects to normal tissues. Chemotherapy-induced toxicities can be long-lasting and may be incompletely reversible; therefore, causative therapies for chemotherapy-dependent side effects are needed, especially considering the increasing survival rates of treated cancer patients. Mesenchymal stem cells (MSCs) have been shown to exhibit regenerative abilities for various forms of tissue damage. Preclinical data suggest that MSCs may also help to alleviate tissue lesions caused by chemotherapeutic agents, mainly by establishing a protective microenvironment for functional cells. Due to the systemic administration of most anticancer agents, the effects of these drugs on the MSCs themselves are of crucial importance to use stem cell-based approaches for the treatment of chemotherapy-induced tissue toxicities. Here, we present a concise review of the published data regarding the influence of various classes of chemotherapeutic agents on the survival, stem cell characteristics and physiological functions of MSCs. Molecular mechanisms underlying the effects are outlined, and resulting challenges of MSC-based treatments for chemotherapy-induced tissue injuries are discussed.
Collapse
Affiliation(s)
- Alexander Rühle
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.,Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Heidelberg, Germany
| | - Peter E Huber
- Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany.,Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Heidelberg, Germany
| | - Rainer Saffrich
- Medical Faculty Mannheim, Institute of Transfusion Medicine and Immunology, German Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany
| | - Ramon Lopez Perez
- Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Heidelberg, Germany
| | - Nils H Nicolay
- Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Heidelberg, Germany.,Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
70
|
Kokturk N, Yıldırım F, Gülhan PY, Oh YM. Stem cell therapy in chronic obstructive pulmonary disease. How far is it to the clinic? AMERICAN JOURNAL OF STEM CELLS 2018; 7:56-71. [PMID: 30245915 PMCID: PMC6146161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 07/01/2018] [Indexed: 06/08/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a respiratory disease that has a major impact worldwide. The currently-available drugs mainly focus on relieving the symptoms of COPD patients. However, in the latter stages of the disease, the airways become largely obstructed and lung parenchyma becomes destructed due to underlying inflammation. The inappropriate repair of lung tissue after injury may contribute to the development of disease. Novel regenerative therapeutic approaches have been investigated with the aim of repairing or replacing the injured functional structures of the respiratory system. Endogenous and exogenous sources of stem cells are available for the treatment of many diseases. Stem cell therapy is newly introduced to the field of COPD. Currently the research is in its infancy; however, the field is profoundly growing. Previous studies suggest that cell-based therapies and novel bioengineering approaches may be potential therapeutic strategies for lung repair and remodelling. In this paper, we review the current evidence of stem cell therapy in COPD.
Collapse
Affiliation(s)
- Nurdan Kokturk
- Department of Pulmonary and Critical Care Medicine, School of Medicine, Gazi UniversityAnkara, Turkey
| | - Fatma Yıldırım
- Dışkapı Yıldırım Beyazıt Research and Education Hospital, Pulmonary & Critical Care MedicineAnkara, Turkey
| | - Pınar Yıldız Gülhan
- Department of Pulmonary Medicine, Düzce University Faculty of MedicineDüzce, Turkey
| | - Yeon Mok Oh
- Department of Pulmonary and Critical Care Medicine and Clinical Research Center for Chronic Obstructive Airway Diseases, Asan Medical Center, University of Ulsan College of MedicineSeoul, South Korea
| |
Collapse
|
71
|
Nunes HC, Scarano WR, Deffune E, Felisbino SL, Porreca I, Delella FK. Bisphenol a and mesenchymal stem cells: Recent insights. Life Sci 2018; 206:22-28. [DOI: 10.1016/j.lfs.2018.05.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 05/07/2018] [Accepted: 05/09/2018] [Indexed: 02/08/2023]
|
72
|
Takeda K, Ning F, Domenico J, Okamoto M, Ashino S, Kim S, Jeong YY, Shiraishi Y, Terada N, Sutherland ER, Gelfand EW. Activation of p70S6 Kinase-1 in Mesenchymal Stem Cells Is Essential to Lung Tissue Repair. Stem Cells Transl Med 2018; 7:551-558. [PMID: 29730892 PMCID: PMC6052610 DOI: 10.1002/sctm.17-0200] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 02/02/2018] [Indexed: 01/06/2023] Open
Abstract
All‐trans retinoic acid (ATRA) or mesenchymal stem cells (MSCs) have been shown to promote lung tissue regeneration in animal models of emphysema. However, the reparative effects of the combination of the two and the role of p70S6 kinase‐1 (p70S6k1) activation in the repair process have not been defined. Twenty‐one days after intratracheal instillation of porcine pancreatic elastase (PPE), MSC and/or 10 days of ATRA treatment was initiated. Thirty‐two days later, static lung compliance (Cst), mean linear intercepts (MLIs), and alveolar surface area (S) were measured. After PPE, mice demonstrated increased values of Cst and MLI, and decreased S values. Both ATRA and MSC transfer were individually effective in improving these outcomes while the combination of ATRA and MSCs was even more effective. The combination of p70S6k1−/− MSCs transfer followed by ATRA demonstrated only modest effects, and rapamycin treatment of recipients with wild‐type (WT) MSCs and ATRA failed to show any effect. However, transfer of p70S6k1 over‐expressing‐MSCs together with ATRA resulted in further improvements over those seen following WT MSCs together with ATRA. ATRA activated p70S6k1 in MSCs in vitro, which was completely inhibited by rapamycin. Tracking of transferred MSCs following ATRA revealed enhanced accumulation and extended survival of MSCs in recipient lungs following PPE but not vehicle instillation. These data suggest that in MSCs, p70S6k1 activation plays a critical role in ATRA‐enhanced lung tissue repair, mediated in part by prolonged survival of transferred MSCs. p70S6k1‐activated MSCs may represent a novel therapeutic approach to reverse the lung damage seen in emphysema. stemcellstranslationalmedicine2018;7:551–558
Collapse
Affiliation(s)
- Katsuyuki Takeda
- Division of Cell Biology, Department of PediatricsNational Jewish HealthDenverColoradoUSA
| | - Fangkun Ning
- Division of Cell Biology, Department of PediatricsNational Jewish HealthDenverColoradoUSA
| | - Joanne Domenico
- Division of Cell Biology, Department of PediatricsNational Jewish HealthDenverColoradoUSA
| | - Masakazu Okamoto
- Division of Cell Biology, Department of PediatricsNational Jewish HealthDenverColoradoUSA
| | - Shigeru Ashino
- Division of Cell Biology, Department of PediatricsNational Jewish HealthDenverColoradoUSA
| | - Sang‐Ha Kim
- Division of Cell Biology, Department of PediatricsNational Jewish HealthDenverColoradoUSA
| | - Yi Yeong Jeong
- Division of Cell Biology, Department of PediatricsNational Jewish HealthDenverColoradoUSA
| | - Yoshiki Shiraishi
- Division of Cell Biology, Department of PediatricsNational Jewish HealthDenverColoradoUSA
| | - Naohiro Terada
- Department of Pathology, Immunology and Laboratory MedicineCollege of Medicine, University of FloridaGainesvilleFloridaUSA
| | | | - Erwin W. Gelfand
- Division of Cell Biology, Department of PediatricsNational Jewish HealthDenverColoradoUSA
| |
Collapse
|
73
|
Kruk DMLW, Heijink IH, Slebos DJ, Timens W, Ten Hacken NH. Mesenchymal Stromal Cells to Regenerate Emphysema: On the Horizon? Respiration 2018; 96:148-158. [PMID: 29719298 DOI: 10.1159/000488149] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 03/02/2018] [Indexed: 12/29/2022] Open
Abstract
Mesenchymal stem or stromal cells (MSCs) are multipotent cells that play a pivotal role in various phases of lung development and lung homeostasis, and potentially also lung regeneration. MSCs do not only self-renew and differentiate into renew tissues, but also have anti-inflammatory and paracrine properties to reduce damage and to support tissue regeneration, constituting a promising cell-based treatment strategy for the repair of damaged alveolar tissue in emphysema. This review discusses the current state of the art regarding the potential of MSCs for the treatment of emphysema. The optimism regarding this treatment strategy is supported by promising results from animal models. Still, there are considerable challenges before effective stem cell treatment can be realized in emphysema patients. It is difficult to draw definitive conclusions from the available animal studies, as different models, dosage protocols, administration routes, and sources of MSCs have been used with different measures of effectiveness. Moreover, the regrowth potential of differentiated tissues and organs differs between species. Essential questions about MSC engraftment, retention, and survival have not been sufficiently addressed in a systematic manner. Few human studies have investigated MSC treatment for chronic obstructive pulmonary disease, demonstrating short-term safety but no convincing benefits on clinical outcomes. Possible explanations for the lack of beneficial effects on clinical outcomes could be the source (bone marrow), route, dosage, frequency of administration, and delivery (lack of a bioactive scaffold). This review will provide a comprehensive overview of the (pre)clinical studies on MSC effects in emphysema and discuss the current challenges regarding the optimal use of MSCs for cell-based therapies.
Collapse
Affiliation(s)
- Dennis M L W Kruk
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Irene H Heijink
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Department of Pulmonary Diseases, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Dirk-Jan Slebos
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Department of Pulmonary Diseases, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Wim Timens
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Department of Pulmonary Diseases, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Nick H Ten Hacken
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.,Department of Pulmonary Diseases, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
74
|
Broekman W, Khedoe PPSJ, Schepers K, Roelofs H, Stolk J, Hiemstra PS. Mesenchymal stromal cells: a novel therapy for the treatment of chronic obstructive pulmonary disease? Thorax 2018; 73:565-574. [PMID: 29653970 PMCID: PMC5969341 DOI: 10.1136/thoraxjnl-2017-210672] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 03/18/2018] [Accepted: 03/26/2018] [Indexed: 12/11/2022]
Abstract
COPD is characterised by tissue destruction and inflammation. Given the lack of curative treatments and the progressive nature of the disease, new treatments for COPD are highly relevant. In vitro cell culture and animal studies have demonstrated that mesenchymal stromal cells (MSCs) have the capacity to modify immune responses and to enhance tissue repair. These properties of MSCs provided a rationale to investigate their potential for treatment of a variety of diseases, including COPD. Preclinical models support the hypothesis that MSCs may have clinical efficacy in COPD. However, although clinical trials have demonstrated the safety of MSC treatment, thus far they have not provided evidence for MSC efficacy in the treatment of COPD. In this review, we discuss the rationale for MSC-based cell therapy in COPD, the main findings from in vitro and in vivo preclinical COPD model studies, clinical trials in patients with COPD and directions for further research.
Collapse
Affiliation(s)
- Winifred Broekman
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Padmini P S J Khedoe
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Koen Schepers
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Helene Roelofs
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Jan Stolk
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
75
|
Lung Mesenchymal Stem Cells Ameliorate Elastase-Induced Damage in an Animal Model of Emphysema. Stem Cells Int 2018; 2018:9492038. [PMID: 29731780 PMCID: PMC5872595 DOI: 10.1155/2018/9492038] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 11/15/2017] [Accepted: 12/21/2017] [Indexed: 12/28/2022] Open
Abstract
Pulmonary emphysema is a respiratory condition characterized by alveolar destruction that leads to airflow limitation and reduced lung function. Although with extensive research, the pathophysiology of emphysema is poorly understood and effective treatments are still missing. Evidence suggests that mesenchymal stem cells (MSCs) possess the ability to engraft the injured tissues and induce repair via a paracrine effect. Thus, the aim of this study was to test the effects of the intratracheal administration of lung-derived mouse MSCs in a model of elastase-induced emphysema. Pulmonary function (static lung compliance) showed an increased stiffness induced by elastase, while morphometric findings (mean linear intercept and tissue/alveolar area) confirmed the severity of alveolar disruption. Contrarily, MSC administration partially restored lung elasticity and alveolar architecture. In the absence of evidence that MSCs acquired epithelial phenotype, we detected an increased proliferative activity of aquaporin 5- and surfactant protein C-positive lung cells, suggesting MSC-driven paracrine mechanisms. The data indicate the mediation of hepatocyte growth factor in amplifying MSC-driven tissue response after injury. Our study shed light on supportive properties of lung-derived MSCs, although the full identification of mechanisms orchestrated by MSCs and responsible for epithelial repair after injury is a critical aspect yet to be achieved.
Collapse
|
76
|
Shafei AES, Ali MA, Ghanem HG, Shehata AI, Abdelgawad AA, Handal HR, ElSayed AS, Ashaal AE, Ali MM, El-Shal AS. Mechanistic effects of mesenchymal and hematopoietic stem cells: New therapeutic targets in myocardial infarction. J Cell Biochem 2018; 119:5274-5286. [PMID: 29266431 DOI: 10.1002/jcb.26637] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Accepted: 12/19/2017] [Indexed: 12/16/2022]
Abstract
Myocardial infarction (MI) results in dysfunction and irreversible loss of cardiomyocytes and is of the most serious health threats today. Mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs) have been explored as promising cell therapy in MI and regenerative therapy. Recently, reports investigated the potential therapeutic effects of MSCs or HSCs transplantation after MI in numerous experimental and clinical studies; however, their results are controversy and needs more explorations. The current review is an attempt to clarify the therapeutic potentials of MSCs and HSCs in MI therapy, as well as their possible effects; especially the paracrine one and the exosome-derived stem cell among animal models as well as clinical trials conducted within the last 10 years. In this context, various sources of MSCs and HSCs have been addressed in helping cardiac regeneration by either revitalizing the cardiac stem cells niche or revascularizing the arteries and veins of the heart. In addition, both MSCs and HSCs could produce paracrine mediators and growth factors which led to cardiomyocytes protection, angiogenesis, immunemodulation, antioxidants, anti-apoptotic, anti-inflammatory, antifibrotic, as well as increasing cardiac contractility. Recently, microRNAs (miRNAs), post-transcriptional regulators of gene expression, and long non-coding RNA (lncRNA), a miRNA sponge, are recent stem cell-derived mediators can be promising targets of MSCs and HSCs through their paracrine effects. Although MSCs and HSCs have achieved considerable achievements, however, some challenges still remain that need to be overcome in order to establish it as a successful technique. The present review clarified the mechanistic potentials of MSCs and HSCs especially paracrine effects involved in MI including human and animal studies and the challenges challenges regarding type, differentiation, route, and number of injections.
Collapse
Affiliation(s)
- Ayman El-Sayed Shafei
- Biomedical Research Department, Military Armed Forces College of Medicine (AFCM), Cairo, Egypt
| | - Mahmoud A Ali
- Biomedical Research Department, Military Armed Forces College of Medicine (AFCM), Cairo, Egypt
| | - Hazem G Ghanem
- Undergraduate Student, Armed Forces College of Medicine, Cairo, Egypt
| | - Ahmed I Shehata
- Undergraduate Student, Armed Forces College of Medicine, Cairo, Egypt
| | | | - Hossam R Handal
- Undergraduate Student, Armed Forces College of Medicine, Cairo, Egypt
| | | | - Ahmed E Ashaal
- Undergraduate Student, Armed Forces College of Medicine, Cairo, Egypt
| | - Mazen M Ali
- Undergraduate Student, Armed Forces College of Medicine, Cairo, Egypt
| | - Amal S El-Shal
- Department of Medical Biochemistry and Molecular biology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
77
|
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: 8] [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
|
78
|
Sun Z, Li F, Zhou X, Chung KF, Wang W, Wang J. Stem cell therapies for chronic obstructive pulmonary disease: current status of pre-clinical studies and clinical trials. J Thorac Dis 2018; 10:1084-1098. [PMID: 29607186 DOI: 10.21037/jtd.2018.01.46] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a respiratory disease that has a major impact worldwide. The currently-available drugs mainly focus on relieving the symptoms of COPD patients. Novel regenerative therapeutic approaches have been investigated with the aim of repairing or replacing the injured functional structures of the respiratory system. We summarized the progress made by regenerative therapies for COPD by analyzing results from both pre-clinical studies and completed clinical trials. These approaches include the application of exogenous stem cells or small molecules to stimulate the regeneration by endogenous lung stem/progenitor cells. Exogenous mesenchymal stem cells (MSCs) have been reported to repair the structure and improve the function of the injured respiratory system in COPD models. However, the studies that used MSCs in patients with moderate-to-severe COPD patients did not lead to clear respiratory functional improvements. Exogenous human lung stem cells applied to cryo-injured (CI) lungs of mice have been shown to organize into human-like pulmonary structures, indicating a new property of stem cells that is potentially capable of curing COPD patients. Small molecules like retinoic acid has been shown to lead to regeneration and repair of the damaged lung structures in COPD mouse models probably by activation of endogenous lung stem/progenitor cells. However, retinoic acid or agonists of retinoic acid receptor administered to moderate or severe COPD patients did not improve the density and function of the damaged lung. These novel regenerative approaches have failed in preliminary clinical trials, possibly due to the advanced severity of the disease. Further work should be done to develop the current regenerative approaches for curing patients at different stages of COPD. We suggest that some modifications of the approach in the clinical studies may lead to more successful outcomes of regenerative therapy for COPD.
Collapse
Affiliation(s)
- Zhongwei Sun
- Cellular Biomedicine Group, Shanghai 200233, China.,Cellular Biomedicine Group, Cupertino, CA, USA
| | - Feng Li
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai 200080, China
| | - Xin Zhou
- Department of Respiratory Medicine, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai 200080, China
| | - Kian Fan Chung
- Airway Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Wen Wang
- Cellular Biomedicine Group, Shanghai 200233, China.,Cellular Biomedicine Group, Cupertino, CA, USA
| | - Jialun Wang
- Cellular Biomedicine Group, Shanghai 200233, China.,Cellular Biomedicine Group, Cupertino, CA, USA
| |
Collapse
|
79
|
Hwang I, Hong S. Neural Stem Cells and Its Derivatives as a New Material for Melanin Inhibition. Int J Mol Sci 2017; 19:ijms19010036. [PMID: 29271951 PMCID: PMC5795986 DOI: 10.3390/ijms19010036] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 12/15/2017] [Accepted: 12/19/2017] [Indexed: 02/07/2023] Open
Abstract
The pigment molecule, melanin, is produced from melanosomes of melanocytes through melanogenesis, which is a complex process involving a combination of chemical and enzymatically catalyzed reactions. The synthesis of melanin is primarily influenced by tyrosinase (TYR), which has attracted interest as a target molecule for the regulation of pigmentation or depigmentation in skin. Thus, direct inhibitors of TYR activity have been sought from various natural and synthetic materials. However, due to issues with these inhibitors, such as weak or permanent ability for depigmentation, allergy, irritant dermatitis and rapid oxidation, in vitro and in vivo, the development of new materials that inhibit melanin production is essential. A conditioned medium (CM) derived from stem cells contains many cell-secreted factors, such as cytokines, chemokines, growth factors and extracellular vesicles including exosomes. In addition, the secreted factors could negatively regulate melanin production through stimulation of a microenvironment of skin tissue in a paracrine manner, which allows the neural stem cell CM to be explored as a new material for skin depigmentation. In this review, we will summarize the current knowledge regulating depigmentation, and discuss the potential of neural stem cells and their derivatives, as a new material for skin depigmentation.
Collapse
Affiliation(s)
- Insik Hwang
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, 22 Gil Inchon-ro, Seongbuk-gu, Seoul 02855, Korea.
- Department of Public Health Sciences, Korea University Graduate School, 22 Gil Inchon-ro, Seongbuk-gu, Seoul 02855, Korea.
| | - Sunghoi Hong
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, 22 Gil Inchon-ro, Seongbuk-gu, Seoul 02855, Korea.
- Department of Public Health Sciences, Korea University Graduate School, 22 Gil Inchon-ro, Seongbuk-gu, Seoul 02855, Korea.
- Department of Integrated Biomedical and Life Science, Korea University Graduate School, 22 Gil Inchon-ro, Seongbuk-gu, Seoul 02855, Korea.
| |
Collapse
|
80
|
Focus on Mesenchymal Stem Cell-Derived Exosomes: Opportunities and Challenges in Cell-Free Therapy. Stem Cells Int 2017; 2017:6305295. [PMID: 29410682 PMCID: PMC5749272 DOI: 10.1155/2017/6305295] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 11/05/2017] [Accepted: 11/22/2017] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stem cells have been at the forefront of regenerative medicine for many years. Exosomes, which are nanovesicles involved in intercellular communication and the transportation of genetic material transportation that can be released by mesenchymal stem cells, have been recently reported to play a role in cell-free therapy of many diseases, including myocardial infarction, drug addiction, and status epilepticus. They are also thought to help ameliorate inflammation-induced preterm brain injury, liver injury, and various types of cancer. This review highlights recent advances in the exploration of mesenchymal stem cell-derived exosomes in therapeutic applications. The natural contents, drug delivery potency, modification methods, and drug loading methods of exosomes are also discussed.
Collapse
|
81
|
Shafei AES, Ali MA, Ghanem HG, Shehata AI, Abdelgawad AA, Handal HR, Talaat KA, Ashaal AE, El-Shal AS. Mesenchymal stem cell therapy: A promising cell-based therapy for treatment of myocardial infarction. J Gene Med 2017; 19. [PMID: 29044850 DOI: 10.1002/jgm.2995] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 10/07/2017] [Accepted: 10/07/2017] [Indexed: 12/12/2022] Open
Abstract
For decades, mesenchymal stem (MSCs) cells have been used for cardiovascular diseases as regenerative therapy. This review is an attempt to summarize the types of MSCs involved in myocardial infarction (MI) therapy, as well as its possible mechanisms effects, especially the paracrine one in MI focusing on the studies (human and animal) conducted within the last 10 years. Recently, reports showed that MSC therapy could have infarct-limiting effects after MI in both experimental and clinical trials. In this context, various types of MSCs can help cardiac regeneration by either revitalizing the cardiac stem cells or revascularizing the arteries and veins of the heart. Furthermore, MSCs could produce paracrine growth factors that increase the survival of nearby cardiomyocytes, as well as increase angiogenesis through recruitment of stem cell from bone marrow or inducing vessel growth from existing capillaries. Recent research suggests that the paracrine effects of MSCs could be mediated by extracellular vesicles including exosomes. Exosomal microRNAs (miRNAs) released by MSCs are promising therapeutic hotspot target for MI. This could be attributed to the role of miRNA in cardiac biology, including cardiac regeneration, stem cell differentiation, apoptosis, neovascularization, cardiac contractility and cardiac remodeling. Furthermore, gene-modified MSCs could be a recent promising therapy for MI to enhance the paracrine effects of MSCs, including better homing and effective cell targeted tissue regeneration. Although MSC therapy has achieved considerable attention and progress, there are critical challenges that remains to be overcome to achieve the most effective successful cell-based therapy in MI.
Collapse
Affiliation(s)
- Ayman El-Sayed Shafei
- Biomedical Research Department, Military Armed Forces College of Medicine, Cairo, Egypt
| | - Mahmoud Ahmed Ali
- Biomedical Research Department, Military Armed Forces College of Medicine, Cairo, Egypt
| | | | | | | | | | | | | | - Amal S El-Shal
- Medical Biochemistry & Molecular Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
82
|
Yudintceva NM, Bogolyubova IO, Muraviov AN, Sheykhov MG, Vinogradova TI, Sokolovich EG, Samusenko IA, Shevtsov MA. Application of the allogenic mesenchymal stem cells in the therapy of the bladder tuberculosis. J Tissue Eng Regen Med 2017; 12:e1580-e1593. [PMID: 28990734 DOI: 10.1002/term.2583] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 09/20/2017] [Accepted: 09/23/2017] [Indexed: 12/26/2022]
Abstract
Urogenital tuberculosis (TB) often leads to contraction of the bladder, a reduction of the urinary reservoir capacity, and, in the latest stage, to real microcystitis up to full obliteration. Bladder TB Stage 4 is unsuitable for conservative therapy, and cystectomy with subsequent enteroplasty is indicated. In this study, using a model of bladder TB in New Zealand rabbits, the therapeutic efficacy of the interstitial injection of autologous bone-derived mesenchymal stem cells (MSCs) combined with standard anti-TB treatment in the restoration of the bladder function was demonstrated. For analysis of the MSC distribution in tissues, the latter were labelled with superparamagnetic iron oxide nanoparticles. In vitro studies demonstrated the high intracellular incorporation of nanoparticles and the absence of cytotoxicity on MSC viability and proliferation. A single-dose administration of MSCs into the bladder mucosal layer significantly reduced the wall deformation and inflammation and hindered the development of fibrosis, which was proven by the subsequent histological assay. Confocal microscopy studies of the bladder cryosections confirmed the presence of superparamagnetic iron oxide nanoparticle-labelled MSCs in different bladder layers of the treated animals, thus indicating the role of stem cells in bladder regeneration.
Collapse
Affiliation(s)
- Natalia M Yudintceva
- Cell Technology Center, Institute of Cytology of the Russian Academy of Sciences (RAS), St. Petersburg, Russia
| | - Irina O Bogolyubova
- Cell Technology Center, Institute of Cytology of the Russian Academy of Sciences (RAS), St. Petersburg, Russia
| | - Alexandr N Muraviov
- Saint-Petersburg State Research Institute of Phthisiopulmonology of the Ministry of Healthcare of the Russian Federation, St. Petersburg, Russia
| | - Magomed G Sheykhov
- Saint-Petersburg State Research Institute of Phthisiopulmonology of the Ministry of Healthcare of the Russian Federation, St. Petersburg, Russia
| | - Tatiana I Vinogradova
- Saint-Petersburg State Research Institute of Phthisiopulmonology of the Ministry of Healthcare of the Russian Federation, St. Petersburg, Russia
| | - Evgenii G Sokolovich
- Saint-Petersburg State Research Institute of Phthisiopulmonology of the Ministry of Healthcare of the Russian Federation, St. Petersburg, Russia.,Saint Petersburg University, St. Petersburg, Russia
| | - Igor A Samusenko
- Federal State Budgetary Institute «The Nikiforov Russian Center of Emergency and Radiation Medicine» (Ministry of Russian Federation for Civil Defense, Emergencies and Elimination of Consequences of Natural Disasters), St. Petersburg, Russia
| | - Maxim A Shevtsov
- Cell Technology Center, Institute of Cytology of the Russian Academy of Sciences (RAS), St. Petersburg, Russia.,First I.P. Pavlov State Medical University of St. Petersburg, St. Petersburg, Russia.,Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| |
Collapse
|
83
|
Janczewski AM, Wojtkiewicz J, Malinowska E, Doboszyńska A. Can Youthful Mesenchymal Stem Cells from Wharton's Jelly Bring a Breath of Fresh Air for COPD? Int J Mol Sci 2017; 18:ijms18112449. [PMID: 29156550 PMCID: PMC5713416 DOI: 10.3390/ijms18112449] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 11/13/2017] [Accepted: 11/14/2017] [Indexed: 12/13/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a major global cause of morbidity and mortality, projected to become the 3rd cause of disease mortality worldwide by 2020. COPD is characterized by persistent and not fully reversible airflow limitation that is usually progressive and is associated with an abnormal chronic inflammatory response of the lung to noxious agents including cigarette smoke. Currently available therapeutic strategies aim to ease COPD symptoms but cannot prevent its progress or regenerate physiological lung structure or function. The urgently needed new approaches for the treatment of COPD include stem cell therapies among which transplantation of mesenchymal stem cells derived from Wharton’s jelly (WJ-MSCs) emerges as a promising therapeutic strategy because of the unique properties of these cells. The present review discusses the main biological properties of WJ-MSCs pertinent to their potential application for the treatment of COPD in the context of COPD pathomechanisms with emphasis on chronic immune inflammatory processes that play key roles in the development and progression of COPD.
Collapse
Affiliation(s)
- Andrzej M Janczewski
- Department of Pulmonology, Faculty of Heath Sciences, University of Warmia and Mazury in Olsztyn, Jagiellońska 78, 10-357 Olsztyn, Poland.
| | - Joanna Wojtkiewicz
- Department of Pathophysiology, Faculty of Medicine, University of Warmia and Mazury in Olsztyn, Warszawska 30, 10-082 Olsztyn, Poland.
- Laboratory for Regenerative Medicine, Faculty of Medicine, University of Warmia and Mazury in Olsztyn, Warszawska 30, 10-082 Olsztyn, Poland.
- Foundation for the Nerve Cells Regeneration, Warszawska 30, 10-082 Olsztyn, Poland.
| | - Ewa Malinowska
- Department of Pulmonology, Faculty of Heath Sciences, University of Warmia and Mazury in Olsztyn, Jagiellońska 78, 10-357 Olsztyn, Poland.
| | - Anna Doboszyńska
- Department of Pulmonology, Faculty of Heath Sciences, University of Warmia and Mazury in Olsztyn, Jagiellońska 78, 10-357 Olsztyn, Poland.
| |
Collapse
|
84
|
Antunes MA, Lapa E Silva JR, Rocco PR. Mesenchymal stromal cell therapy in COPD: from bench to bedside. Int J Chron Obstruct Pulmon Dis 2017; 12:3017-3027. [PMID: 29081655 PMCID: PMC5652911 DOI: 10.2147/copd.s146671] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
COPD is the most frequent chronic respiratory disease and a leading cause of morbidity and mortality. The major risk factor for COPD development is cigarette smoke, and the most efficient treatment for COPD is smoking cessation. However, even after smoking cessation, inflammation, apoptosis, and oxidative stress may persist and continue contributing to disease progression. Although current therapies for COPD (primarily based on anti-inflammatory agents) contribute to the reduction of airway obstruction and minimize COPD exacerbations, none can avoid disease progression or reduce mortality. Within this context, recent advances in mesenchymal stromal cell (MSC) therapy have made this approach a strong candidate for clinical use in the treatment of several pulmonary diseases. MSCs can be readily harvested from diverse tissues and expanded with high efficiency, and have strong immunosuppressive properties. Preclinical studies have demonstrated encouraging outcomes of MSCs therapy for lung disorders, including emphysema. These findings instigated research groups to assess the impact of MSCs in human COPD/emphysema, but clinical results have fallen short of expectations. However, MSCs have demonstrated a good adjuvant role in the clinical scenario. Trials that used MSCs combined with another, primary treatment (eg, endobronchial valves) found that patients derived greater benefit in pulmonary function tests and/or quality of life reports, as well as reductions in systemic markers of inflammation. The present review summarizes and describes the more recent preclinical studies that have been published about MSC therapy for COPD/emphysema and discusses what has already been applied about MSCs treatment in COPD patients in the clinical setting.
Collapse
Affiliation(s)
- Mariana A Antunes
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro (UFRJ), RJ, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ, Brazil
| | - José Roberto Lapa E Silva
- Institute of Thoracic Medicine, Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Patricia Rm Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro (UFRJ), RJ, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
85
|
Khedoe PPSJ, de Kleijn S, van Oeveren-Rietdijk AM, Plomp JJ, de Boer HC, van Pel M, Rensen PCN, Berbée JFP, Hiemstra PS. Acute and chronic effects of treatment with mesenchymal stromal cells on LPS-induced pulmonary inflammation, emphysema and atherosclerosis development. PLoS One 2017; 12:e0183741. [PMID: 28910300 PMCID: PMC5598950 DOI: 10.1371/journal.pone.0183741] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 08/10/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND COPD is a pulmonary disorder often accompanied by cardiovascular disease (CVD), and current treatment of this comorbidity is suboptimal. Systemic inflammation in COPD triggered by smoke and microbial exposure is suggested to link COPD and CVD. Mesenchymal stromal cells (MSC) possess anti-inflammatory capacities and MSC treatment is considered an attractive treatment option for various chronic inflammatory diseases. Therefore, we investigated the immunomodulatory properties of MSC in an acute and chronic model of lipopolysaccharide (LPS)-induced inflammation, emphysema and atherosclerosis development in APOE*3-Leiden (E3L) mice. METHODS Hyperlipidemic E3L mice were intranasally instilled with 10 μg LPS or vehicle twice in an acute 4-day study, or twice weekly during 20 weeks Western-type diet feeding in a chronic study. Mice received 0.5x106 MSC or vehicle intravenously twice after the first LPS instillation (acute study) or in week 14, 16, 18 and 20 (chronic study). Inflammatory parameters were measured in bronchoalveolar lavage (BAL) and lung tissue. Emphysema, pulmonary inflammation and atherosclerosis were assessed in the chronic study. RESULTS In the acute study, intranasal LPS administration induced a marked systemic IL-6 response on day 3, which was inhibited after MSC treatment. Furthermore, MSC treatment reduced LPS-induced total cell count in BAL due to reduced neutrophil numbers. In the chronic study, LPS increased emphysema but did not aggravate atherosclerosis. Emphysema and atherosclerosis development were unaffected after MSC treatment. CONCLUSION These data show that MSC inhibit LPS-induced pulmonary and systemic inflammation in the acute study, whereas MSC treatment had no effect on inflammation, emphysema and atherosclerosis development in the chronic study.
Collapse
Affiliation(s)
- P. Padmini S. J. Khedoe
- Dept. of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
- Dept. of Medicine, Div. of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
| | - Stan de Kleijn
- Dept. of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Annemarie M. van Oeveren-Rietdijk
- Dept. of Medicine, Div. of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Jaap J. Plomp
- Dept. of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hetty C. de Boer
- Dept. of Medicine, Div. of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Melissa van Pel
- Dept. of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Patrick C. N. Rensen
- Dept. of Medicine, Div. of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Jimmy F. P. Berbée
- Dept. of Medicine, Div. of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Pieter S. Hiemstra
- Dept. of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
86
|
Scott A, Glasgow A, Small D, Carlile S, McCrudden M, McLean D, Brown R, Doherty D, Lundy FT, Hamid UI, O'Kane CM, McAuley DF, Brodlie M, Tunney M, Elborn JS, Irwin CR, Timson DJ, Taggart CC, Weldon S. Characterisation of eppin function: expression and activity in the lung. Eur Respir J 2017; 50:50/1/1601937. [DOI: 10.1183/13993003.01937-2016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 03/29/2017] [Indexed: 11/05/2022]
Abstract
Eppin is a serine protease inhibitor expressed in male reproductive tissues.The aim of this study was to investigate the localisation and regulation of eppin expression in myeloid and epithelial cell lines, and explore its potential role as a multifunctional host defence protein.Using immunohistochemistry and Western blotting, eppin was detected in the lungs of patients with acute respiratory distress syndrome and cystic fibrosis lung disease. Expression of eppin in monocytic cells was unaffected by stimulation with Toll-like receptor agonists, cytokines and hormone receptor agonists. However, upregulated expression and secretion of eppin was observed following treatment of monocytes with epidermal growth factor. Incubation of recombinant eppin with monocytic cells resulted in significant inhibition of lipopolysaccharide-induced chemokine production. Furthermore, eppin inhibited lipopolysaccharide-induced NF-κB activation by a mechanism which involved accumulation of phosphorylated IκBα. In anin vivomodel of lung inflammation induced by lipopolysaccharide, eppin administration resulted in decreased recruitment of neutrophils to the lung with a concomitant reduction in the levels of the neutrophil chemokine macrophage inflammatory protein-2.Overall, these results suggest a role for eppin outside of the reproductive tract and that eppin may have a role in the innate immune response in the lung.
Collapse
|
87
|
Lu Z, Chen Y, Dunstan C, Roohani-Esfahani S, Zreiqat H. Priming Adipose Stem Cells with Tumor Necrosis Factor-Alpha Preconditioning Potentiates Their Exosome Efficacy for Bone Regeneration. Tissue Eng Part A 2017; 23:1212-1220. [PMID: 28346798 DOI: 10.1089/ten.tea.2016.0548] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been widely used for tissue repair and regeneration. However, the inherent drawbacks, including limited cell survival after cell transplantation, have hindered direct MSC transplantation for tissue repair and regeneration. The aim of this study was to investigate if exosomes isolated from MSCs can promote the proliferation and differentiation of human primary osteoblastic cells (HOBs) and be potentially used for bone tissue regeneration. We showed that adipose tissue-derived MSC (ASC)-derived exosomes (ASC-EXO) were able to promote the proliferation and osteogenic differentiation in HOBs; and the trophic effects of ASC-EXO on HOBs were further harnessed when ASCs were preconditioned with tumor necrosis factor-alpha (TNF-α) for 3 days, which mimics the acute inflammatory phase upon bone injury. In addition, we showed that Wnt-3a content was elevated in ASC-EXO when ASCs were preconditioned by TNF-α, and inhibiting Wnt signaling decreased the osteogenic gene expression levels in HOBs which were cultured in TNF-α preconditioned ASCs conditioned medium. In conclusion, it was demonstrated that ASC-EXO, especially primed by TNF-α preconditioning on ASCs, offer a promising approach to replace direct stem cell transplantation for bone repair and regeneration.
Collapse
Affiliation(s)
- ZuFu Lu
- Biomaterials and Tissue Engineering Research Unit, School of AMME, The University of Sydney , Sydney, Australia
| | - YongJuan Chen
- Biomaterials and Tissue Engineering Research Unit, School of AMME, The University of Sydney , Sydney, Australia
| | - Colin Dunstan
- Biomaterials and Tissue Engineering Research Unit, School of AMME, The University of Sydney , Sydney, Australia
| | - Seyediman Roohani-Esfahani
- Biomaterials and Tissue Engineering Research Unit, School of AMME, The University of Sydney , Sydney, Australia
| | - Hala Zreiqat
- Biomaterials and Tissue Engineering Research Unit, School of AMME, The University of Sydney , Sydney, Australia
| |
Collapse
|
88
|
Mesenchymal Stem Cell Administration in Patients with Chronic Obstructive Pulmonary Disease: State of the Science. Stem Cells Int 2017; 2017:8916570. [PMID: 28303154 PMCID: PMC5337878 DOI: 10.1155/2017/8916570] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 01/22/2017] [Indexed: 01/10/2023] Open
Abstract
Patients with chronic obstructive pulmonary disease (COPD) have chronic, irreversible airway inflammation; currently, there is no effective or curative treatment and the main goals of COPD management are to mitigate symptoms and improve patients' quality of life. Stem cell based therapy offers a promising therapeutic approach that has shown potential in diverse degenerative lung diseases. Preclinical studies have demonstrated encouraging outcomes of mesenchymal stem/stromal cells (MSCs) therapy for lung disorders including emphysema, bronchopulmonary dysplasia, fibrosis, and acute respiratory distress syndrome. This review summarizes available data on 15 studies currently registered by the ClinicalTrials.gov repository, which used different stem cell therapy protocols for COPD; these included bone marrow mononuclear cells (BMMCs), bone marrow-derived MSCs, adipose-derived stem/stromal cells (ADSCs), and adipose-derived MSCs. Published results of three trials indicate that administering BMMCs or MSCs in the setting of degenerative lung disease is safe and may improve patients' condition and quality of life; however, larger-scale studies are needed to evaluate efficacy. Results of another completed trial (NCT01872624) are not yet published, and eleven other studies are ongoing; these include MSCs therapy in emphysema, several studies of ADSCs in COPD, another in idiopathic pulmonary fibrosis, and plerixafor mobilization of CD117 stem cells to peripheral blood.
Collapse
|
89
|
Adipose stem cell-derived nanovesicles inhibit emphysema primarily via an FGF2-dependent pathway. Exp Mol Med 2017; 49:e284. [PMID: 28082743 PMCID: PMC5291836 DOI: 10.1038/emm.2016.127] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 08/10/2016] [Accepted: 08/18/2016] [Indexed: 12/23/2022] Open
Abstract
Cell therapy using stem cells has produced therapeutic benefits in animal models of COPD. Secretory mediators are proposed as one mechanism for stem cell effects because very few stem cells engraft after injection into recipient animals. Recently, nanovesicles that overcome the disadvantages of natural exosomes have been generated artificially from cells. We generated artificial nanovesicles from adipose-derived stem cells (ASCs) using sequential penetration through polycarbonate membranes. ASC-derived artificial nanovesicles displayed a 100 nm-sized spherical shape similar to ASC-derived natural exosomes and expressed both exosomal and stem cell markers. The proliferation rate of lung epithelial cells was increased in cells treated with ASC-derived artificial nanovesicles compared with cells treated with ASC-derived natural exosomes. The lower dose of ASC-derived artificial nanovesicles had similar regenerative capacity compared with a higher dose of ASCs and ASC-derived natural exosomes. In addition, FGF2 levels in the lungs of mice treated with ASC-derived artificial nanovesicles were increased. The uptake of ASC-derived artificial nanovesicles was inhibited by heparin, which is a competitive inhibitor of heparan sulfate proteoglycan that is associated with FGF2 signaling. Taken together, the data indicate that lower doses of ASC-derived artificial nanovesicles may have beneficial effects similar to higher doses of ASCs or ASC-derived natural exosomes in an animal model with emphysema, suggesting that artificial nanovesicles may have economic advantages that warrant future clinical studies.
Collapse
|
90
|
Oh DK, Kim YS, Oh YM. Lung Regeneration Therapy for Chronic Obstructive Pulmonary Disease. Tuberc Respir Dis (Seoul) 2016; 80:1-10. [PMID: 28119741 PMCID: PMC5256352 DOI: 10.4046/trd.2017.80.1.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 01/13/2016] [Accepted: 07/05/2016] [Indexed: 12/16/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a critical condition with high morbidity and mortality. Although several medications are available, there are no definite treatments. However, recent advances in the understanding of stem and progenitor cells in the lung, and molecular changes during re-alveolization after pneumonectomy, have made it possible to envisage the regeneration of damaged lungs. With this background, numerous studies of stem cells and various stimulatory molecules have been undertaken, to try and regenerate destroyed lungs in animal models of COPD. Both the cell and drug therapies show promising results. However, in contrast to the successes in laboratories, no clinical trials have exhibited satisfactory efficacy, although they were generally safe and tolerable. In this article, we review the previous experimental and clinical trials, and summarize the recent advances in lung regeneration therapy for COPD. Furthermore, we discuss the current limitations and future perspectives of this emerging field.
Collapse
Affiliation(s)
- Dong Kyu Oh
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, Seoul, Korea
| | - You-Sun Kim
- Asan Institute for Life Sciences, Seoul, Korea.; University of Ulsan College of Medicine, Seoul, Korea
| | - Yeon-Mok Oh
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, Seoul, Korea.; Asan Institute for Life Sciences, Seoul, Korea.; University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
91
|
Human mesenchymal stromal cells exert HGF dependent cytoprotective effects in a human relevant pre-clinical model of COPD. Sci Rep 2016; 6:38207. [PMID: 27922052 PMCID: PMC5138599 DOI: 10.1038/srep38207] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 11/07/2016] [Indexed: 12/13/2022] Open
Abstract
Bone-marrow derived mesenchymal stromal cells (MSCs) have potent immunomodulatory and tissue reparative properties, which may be beneficial in the treatment of inflammatory diseases such as COPD. This study examined the mechanisms by which human MSCs protect against elastase induced emphysema. Using a novel human relevant pre-clinical model of emphysema the efficacy of human MSC therapy and optimal cell dose were investigated. Protective effects were examined in the lung through histological examination. Further in vivo experiments examined the reparative abilities of MSCs after tissue damage was established and the role played by soluble factors secreted by MSCs. The mechanism of MSC action was determined in using shRNA gene knockdown. Human MSC therapy and MSC conditioned media exerted significant cytoprotective effects when administered early at the onset of the disease. These protective effects were due to significant anti-inflammatory, anti-fibrotic and anti-apoptotic mechanisms, mediated in part through MSC production of hepatocyte growth factor (HGF). When MSC administration was delayed, significant protection of the lung architecture was observed but this was less extensive. MSC cell therapy was more effective than MSC conditioned medium in this emphysema model.
Collapse
|
92
|
Wang LT, Ting CH, Yen ML, Liu KJ, Sytwu HK, Wu KK, Yen BL. Human mesenchymal stem cells (MSCs) for treatment towards immune- and inflammation-mediated diseases: review of current clinical trials. J Biomed Sci 2016; 23:76. [PMID: 27809910 PMCID: PMC5095977 DOI: 10.1186/s12929-016-0289-5] [Citation(s) in RCA: 231] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 10/12/2016] [Indexed: 12/19/2022] Open
Abstract
Human mesenchymal stem cells (MSCs) are multilineage somatic progenitor/stem cells that have been shown to possess immunomodulatory properties in recent years. Initially met with much skepticism, MSC immunomodulation has now been well reproduced across tissue sources and species to be clinically relevant. This has opened up the use of these versatile cells for application as 3rd party/allogeneic use in cell replacement/tissue regeneration, as well as for immune- and inflammation-mediated disease entities. Most surprisingly, use of MSCs for in immune-/inflammation-mediated diseases appears to yield more efficacy than for regenerative medicine, since engraftment of the exogenous cell does not appear necessary. In this review, we focus on this non-traditional clinical use of a tissue-specific stem cell, and highlight important findings and trends in this exciting area of stem cell therapy.
Collapse
Affiliation(s)
- Li-Tzu Wang
- Regenerative Medicine Research Group, Institute of Cellular & System Medicine, National Health Research Institutes (NHRI), 35 Keyan Road, Zhunan, 35053, Taiwan.,Graduate Institute of Life Sciences, National Defense Medical Center (NDMC), Taipei, Taiwan
| | - Chiao-Hsuan Ting
- Regenerative Medicine Research Group, Institute of Cellular & System Medicine, National Health Research Institutes (NHRI), 35 Keyan Road, Zhunan, 35053, Taiwan
| | - Men-Luh Yen
- Department of Ob/Gyn, National Taiwan University Hospital & College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ko-Jiunn Liu
- National Institute of Cancer Research, NHRI, Tainan, Taiwan
| | - Huey-Kang Sytwu
- Graduate Institute of Life Sciences, National Defense Medical Center (NDMC), Taipei, Taiwan.,Graduate Institute of Microbiology and Immunology, NDMC, Taipei, Taiwan
| | - Kenneth K Wu
- Regenerative Medicine Research Group, Institute of Cellular & System Medicine, National Health Research Institutes (NHRI), 35 Keyan Road, Zhunan, 35053, Taiwan.,Graduate Institute of Basic Medical Sciences, China Medical College, Taichung, Taiwan
| | - B Linju Yen
- Regenerative Medicine Research Group, Institute of Cellular & System Medicine, National Health Research Institutes (NHRI), 35 Keyan Road, Zhunan, 35053, Taiwan.
| |
Collapse
|
93
|
Hong Y, Kim YS, Hong SH, Oh YM. Therapeutic effects of adipose-derived stem cells pretreated with pioglitazone in an emphysema mouse model. Exp Mol Med 2016; 48:e266. [PMID: 27765950 PMCID: PMC5099424 DOI: 10.1038/emm.2016.93] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 05/17/2016] [Accepted: 05/19/2016] [Indexed: 12/16/2022] Open
Abstract
There is no therapy currently available that influences the natural history of disease progression in patients with chronic obstructive pulmonary disease (COPD). Although stem cell therapy is considered a potential therapeutic option in COPD, there are no clinical trials proving definitive therapeutic effects in patients with COPD. Recently, it was reported that pioglitazone might potentiate the therapeutic effects of stem cells in patients with heart or liver disease. To test the capacity of pioglitazone pretreatment of stem cells for emphysema repair, we evaluated the therapeutic effects of pioglitazone-pretreated human adipose-derived mesenchymal stem cells (ASCs) on elastase-induced or cigarette smoke-induced emphysema in mice. We also investigated the mechanisms of action of pioglitazone-pretreated ASCs. Pioglitazone-pretreated ASCs had a more potent therapeutic effect than non-pretreated ASCs in the repair of both elastase-induced and smoke-induced emphysema models (mean linear intercept, 78.1±2.5 μm vs 83.2±2.6 μm in elastase models and 75.6±1.4 μm vs 80.5±3.2 μm in smoke models, P<0.05). Furthermore, we showed that pioglitazone-pretreated ASCs increased vascular endothelial growth factor (VEGF) production both in vitro and in mouse lungs in the smoke-induced emphysema model. Pioglitazone-pretreated ASCs may have more potent therapeutic effects than non-pretreated ASCs in emphysema mouse models.
Collapse
Affiliation(s)
- Yoonki Hong
- Department of Internal Medicine, Kangwon National University Hospital, Chuncheon, Korea
| | - You-Sun Kim
- Asan Institute for Life Sciences, Department of Pulmonary and Critical Care Medicine, University of Ulsan College of Medicine, Seoul, Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, Kangwon National University Hospital, Chuncheon, Korea
| | - Yeon-Mok Oh
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, Seoul, Korea
| |
Collapse
|
94
|
Kim YS, Kokturk N, Kim JY, Lee SW, Lim J, Choi SJ, Oh W, Oh YM. Gene Profiles in a Smoke-Induced COPD Mouse Lung Model Following Treatment with Mesenchymal Stem Cells. Mol Cells 2016; 39:728-733. [PMID: 27802588 PMCID: PMC5104880 DOI: 10.14348/molcells.2016.0095] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 09/07/2016] [Accepted: 09/07/2016] [Indexed: 01/08/2023] Open
Abstract
Mesenchymal stem cells (MSCs) effectively reduce airway inflammation and regenerate the alveolus in cigarette- and elastase-induced chronic obstructive pulmonary disease (COPD) animal models. The effects of stem cells are thought to be paracrine and immune-modulatory because very few stem cells remain in the lung one day after their systemic injection, which has been demonstrated previously. In this report, we analyzed the gene expression profiles to compare mouse lungs with chronic exposure to cigarette smoke with non-exposed lungs. Gene expression profiling was also conducted in a mouse lung tissue with chronic exposure to cigarette smoke following the systemic injection of human cord blood-derived mesenchymal stem cells (hCB-MSCs). Globally, 834 genes were differentially expressed after systemic injection of hCB-MSCs. Seven and 21 genes, respectively, were up-and downregulated on days 1, 4, and 14 after HCB-MSC injection. The Hbb and Hba, genes with oxygen transport and antioxidant functions, were increased on days 1 and 14. A serine protease inhibitor was also increased at a similar time point after injection of hCB-MSCs. Gene Ontology analysis indicated that the levels of genes related to immune responses, metabolic processes, and blood vessel development were altered, indicating host responses after hCB-MSC injection. These gene expression changes suggest that MSCs induce a regeneration mechanism against COPD induced by cigarette smoke. These analyses provide basic data for understanding the regeneration mechanisms promoted by hCB-MSCs in cigarette smoke-induced COPD.
Collapse
Affiliation(s)
- You-Sun Kim
- University of Ulsan College of Medicine, Seoul 05505,
Korea
- Asan Institute for Life Sciences, Seoul 05505,
Korea
| | - Nurdan Kokturk
- Department of Pulmonology, Gazi University, Ankara,
Turkey
| | - Ji-Young Kim
- Asan Institute for Life Sciences, Seoul 05505,
Korea
| | - Sei Won Lee
- University of Ulsan College of Medicine, Seoul 05505,
Korea
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, Seoul 05505,
Korea
| | - Jaeyun Lim
- University of Ulsan College of Medicine, Seoul 05505,
Korea
| | - Soo Jin Choi
- Biomedical Research Institute, MEDIPOST Co., Ltd., Seoul,
Korea
| | - Wonil Oh
- Biomedical Research Institute, MEDIPOST Co., Ltd., Seoul,
Korea
| | - Yeon-Mok Oh
- University of Ulsan College of Medicine, Seoul 05505,
Korea
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, Seoul 05505,
Korea
| |
Collapse
|
95
|
Nong K, Wang W, Niu X, Hu B, Ma C, Bai Y, Wu B, Wang Y, Ai K. Hepatoprotective effect of exosomes from human-induced pluripotent stem cell-derived mesenchymal stromal cells against hepatic ischemia-reperfusion injury in rats. Cytotherapy 2016; 18:1548-1559. [PMID: 27592404 DOI: 10.1016/j.jcyt.2016.08.002] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 08/01/2016] [Accepted: 08/07/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND This study aimed to evaluate the effect of exosomes produced by human-induced pluripotent stem cell-derived mesenchymal stromal cells (hiPSC-MSCs-Exo) on hepatic ischemia-reperfusion (I/R) injury. METHODS Exosomes were isolated and concentrated from conditioned medium using ultracentrifugation and ultrafiltration. hiPSC-MSCs-Exo were injected systemically via the inferior vena cava in a rat model of 70% warm hepatic I/R injury, and the therapeutic effect was evaluated. The serum levels of transaminases (aspartate aminotransferase [AST] and alanine aminotransferase [ALT]) were measured using an automatic analyzer. The expression of inflammatory factors was measured using enzyme-linked immunosorbent assay (ELISA). Histological changes indicated changes in pathology and inflammatory infiltration in liver tissue. Apoptosis of hepatic cells in liver tissue was measured using terminal-deoxynucleoitidyl transferase mediated nick end labeling (TUNEL) staining along with apoptotic markers. RESULTS hiPSCs were efficiently induced into hiPSC-MSCs with typical MSC characteristics. hiPSC-MSCs-Exo had diameters ranging from 50 to 60 nm and expressed exosomal markers (CD9, CD63 and CD81). Hepatocyte necrosis and sinusoidal congestion were markedly suppressed with a lower Suzuki score after hiPSC-MSCs-Exo administration. The levels of the hepatocyte injury markers AST and ALT were significantly lower in the treated group than in the control group. Inflammatory markers, such as tumor necrosis factor (TNF)-α, interleukin (IL)-6 and high mobility group box 1 (HMGB1), were significantly reduced after administration of hiPSC-MSCs-Exo, which suggests that the exosomes have a role in suppressing the inflammatory response. Additionally, in liver tissues from the experimental group, the levels of apoptotic markers, such as caspase-3 and bax, were significantly lower and the levels of oxidative markers, such as glutathione (GSH), glutathione peroxidase (GSH-Px) and superoxide dismutase (SOD), were significantly higher than in the control group. These data point to an anti-apoptotic, anti-oxidative stress response role for hiPSC-MSCs-Exo. CONCLUSIONS Our results demonstrated that hiPSC-MSCs-Exo alleviate hepatic I/R injury, possibly via suppression of inflammatory responses, attenuation of the oxidative stress response and inhibition of apoptosis.
Collapse
Affiliation(s)
- Kate Nong
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Weiwei Wang
- Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Xin Niu
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Bin Hu
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Chenchao Ma
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yueqing Bai
- Department of Pathology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Bo Wu
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yang Wang
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| | - Kaixing Ai
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China; Tongji Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
96
|
Wen D, Peng Y, Liu D, Weizmann Y, Mahato RI. Mesenchymal stem cell and derived exosome as small RNA carrier and Immunomodulator to improve islet transplantation. J Control Release 2016; 238:166-175. [PMID: 27475298 DOI: 10.1016/j.jconrel.2016.07.044] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 07/20/2016] [Accepted: 07/25/2016] [Indexed: 01/01/2023]
Abstract
Human bone marrow mesenchymal stem cells (hBMSCs) and their exosomes can suppress immune reaction and deliver small RNAs. Thus, they may improve islet transplantation by delivering small RNAs for promoting islet function and inhibiting immune rejection. Here, we proposed an hBMSC and its exosome-based therapy to overcome immune rejection and poor islet function, both of which hinder the success of islet transplantation. We found overexpressed siFas and anti-miR-375 in plasmid encoding shFas and anti-miR-375 transfected hBMSC-derived exosomes, which silenced Fas and miR-375 of human islets and improved their viability and function against inflammatory cytokines. This plasmid transfected hBMSCs downregulated Fas and miR-375 of human islets in a humanized NOD scid gamma (NSG) mouse model, whose immune reaction was inhibited by injecting hBMSC and peripheral blood mononuclear cell (PBMC) co-cultured exosomes. These exosomes suppressed immune reaction by inhibiting PBMC proliferation and enhancing regulatory T cell (Treg) function. Collectively, our studies elucidated the mechanisms of RNA delivery from hBMSCs to human islets and the immunosuppressive effect of hBMSC and peripheral blood mononuclear cell co-cultured exosomes for improving islet transplantation.
Collapse
Affiliation(s)
- Di Wen
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Yang Peng
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Di Liu
- Department of Chemistry, University of Chicago, Chicago, IL 60637, United States
| | - Yossi Weizmann
- Department of Chemistry, University of Chicago, Chicago, IL 60637, United States
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States.
| |
Collapse
|
97
|
Liu X, Fang Q, Kim H. Preclinical Studies of Mesenchymal Stem Cell (MSC) Administration in Chronic Obstructive Pulmonary Disease (COPD): A Systematic Review and Meta-Analysis. PLoS One 2016; 11:e0157099. [PMID: 27280283 PMCID: PMC4900582 DOI: 10.1371/journal.pone.0157099] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 05/24/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND In the last two decades, mesenchymal stem cells (MSCs) have been pre-clinically utilized in the treatment of a variety of kinds of diseases including chronic obstructive pulmonary disease (COPD). The aim of the current study was to systematically review and conduct a meta-analysis on the published pre-clinical studies of MSC administration in the treatment of COPD in animal models. METHODS AND RESULTS A systematic search of electronic databases was performed. Statistical analysis was performed using the Comprehensive Meta-Analysis software (Version 3). The pooled Hedges's g with 95% confidence intervals (95% CIs) was adopted to assess the effect size. Random effect model was used due to the heterogeneity between the studies. A total of 20 eligible studies were included in the current systematic review. The overall meta-analysis showed that MSC administration was significantly in favor of attenuating acute lung injury (Hedges's g = -2.325 ± 0.145 with 95% CI: -2.609 ~ -2.040, P < 0.001 for mean linear intercept, MLI; Hedges's g = -3.488 ± 0.504 with 95% CI: -4.476 ~ -2.501, P < 0.001 for TUNEL staining), stimulating lung tissue repair (Hedges's g = 3.249 ± 0.586 with 95% CI: 2.103~ 4.394, P < 0.001) and improving lung function (Hedges's g = 2.053 ± 0.408 with 95% CI: 1.253 ~ 2.854, P< 0.001). The mechanism of MSC therapy in COPD is through ameliorating airway inflammation (Hedges's g = -2.956 ± 0.371 with 95% CI: -3.683 ~ -2.229, P< 0.001) and stimulating cytokine synthesis that involves lung tissue repair (Hedges's g = 3.103 ± 0.734 with 95% CI: 1.664 ~ 4.541, P< 0.001). CONCLUSION This systematic review and meta-analysis suggest a promising role for MSCs in COPD treatment. Although the COPD models may not truly mimic COPD patients, these pre-clinical studies demonstrate that MSC hold promise in the treatment of chronic lung diseases including COPD. The mechanisms of MSCs role in preclinical COPD treatment may be associated with attenuating airway inflammation as well as stimulating lung tissue repair.
Collapse
Affiliation(s)
- Xiangde Liu
- Pulmonary, Critical Care, Sleep and Allergy Medicine, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Qiuhong Fang
- Department of Pulmonary and Critical Care, Beijing Chaoyang Hospital, The Capital Medical University, Beijing, China
| | - Huijung Kim
- Pulmonary and Critical Care Division, WonKwang University, Sanbon Medical Center, Seoul, Korea
- * E-mail:
| |
Collapse
|
98
|
Laube M, Stolzing A, Thome UH, Fabian C. Therapeutic potential of mesenchymal stem cells for pulmonary complications associated with preterm birth. Int J Biochem Cell Biol 2016; 74:18-32. [PMID: 26928452 DOI: 10.1016/j.biocel.2016.02.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 02/23/2016] [Accepted: 02/25/2016] [Indexed: 12/22/2022]
Abstract
Preterm infants frequently suffer from pulmonary complications resulting in significant morbidity and mortality. Physiological and structural lung immaturity impairs perinatal lung transition to air breathing resulting in respiratory distress. Mechanical ventilation and oxygen supplementation ensure sufficient oxygen supply but enhance inflammatory processes which might lead to the establishment of a chronic lung disease called bronchopulmonary dysplasia (BPD). Current therapeutic options to prevent or treat BPD are limited and have salient side effects, highlighting the need for new therapeutic approaches. Mesenchymal stem cells (MSCs) have demonstrated therapeutic potential in animal models of BPD. This review focuses on MSC-based therapeutic approaches to treat pulmonary complications and critically compares results obtained in BPD models. Thereby bottlenecks in the translational systems are identified that are preventing progress in combating BPD. Notably, current animal models closely resemble the so-called "old" BPD with profound inflammation and injury, whereas clinical improvements shifted disease pathology towards a "new" BPD in which arrest of lung maturation predominates. Future studies need to evaluate the utility of MSC-based therapies in animal models resembling the "new" BPD though promising in vitro evidence suggests that MSCs do possess the potential to stimulate lung maturation. Furthermore, we address the mode-of-action of MSC-based therapies with regard to lung development and inflammation/fibrosis. Their therapeutic efficacy is mainly attributed to an enhancement of regeneration and immunomodulation due to paracrine effects. In addition, we discuss current improvement strategies by genetic modifications or precondition of MSCs to enhance their therapeutic efficacy which could also prove beneficial for BPD therapies.
Collapse
Affiliation(s)
- Mandy Laube
- Center for Pediatric Research Leipzig, Hospital for Children & Adolescents, Division of Neonatology, University of Leipzig, Leipzig, Germany.
| | - Alexandra Stolzing
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany; Loughborough University, Wolfson School of Mechanical and Manufacturing Engineering, Centre for Biological Engineering, Loughborough, UK.
| | - Ulrich H Thome
- Center for Pediatric Research Leipzig, Hospital for Children & Adolescents, Division of Neonatology, University of Leipzig, Leipzig, Germany.
| | - Claire Fabian
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany; Interdisciplinary Centre for Bioinformatics, University of Leipzig, Leipzig, Germany.
| |
Collapse
|
99
|
Savukinas UB, Enes SR, Sjöland AA, Westergren-Thorsson G. Concise Review: The Bystander Effect: Mesenchymal Stem Cell-Mediated Lung Repair. Stem Cells 2016; 34:1437-44. [PMID: 26991735 DOI: 10.1002/stem.2357] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 02/15/2016] [Indexed: 12/15/2022]
Abstract
Mesenchymal stem or stromal cells (MSCs), a heterogeneous subset of adult stem/progenitor cells, have surfaced as potential therapeutic units with significant clinical benefit for a wide spectrum of disease conditions, including those affecting the lung. Although MSCs carry both self-renewal and multilineage differentiation abilities, current dogma holds that MSCs mainly contribute to tissue regeneration and repair by modulating the host tissue via secreted cues. Thus, the therapeutic benefit of MSCs is thought to derive from so called bystander effects. The regenerative mechanisms employed by MSCs in the lung include modulation of the immune system as well as promotion of epithelial and endothelial repair. Apart from secreted factors, a number of recent findings suggest that MSCs engage in mitochondrial transfer and shedding of membrane vesicles as a means to enhance tissue repair following injury. Furthermore, it is becoming increasingly clear that MSCs are an integral component of epithelial lung stem cell niches. As such, MSCs play an important role in coupling information from the environment to stem and progenitor populations, such that homeostasis can be ensured even in the face of injury. It is the aim of this review to outline the major mechanisms by which MSCs contribute to lung regeneration, synthesizing recent preclinical findings with data from clinical trials and potential for future therapy. Stem Cells 2016;34:1437-1444.
Collapse
Affiliation(s)
- Ulrika Blank Savukinas
- Department of Experimental Medical Science, Lung Biology Unit, Lund University, Lund, Sweden
| | - Sara Rolandsson Enes
- Department of Experimental Medical Science, Lung Biology Unit, Lund University, Lund, Sweden
| | | | | |
Collapse
|
100
|
Dendrobium officinale polysaccharides ameliorated pulmonary function while inhibiting mucin-5AC and stimulating aquaporin-5 expression. J Funct Foods 2016. [DOI: 10.1016/j.jff.2015.12.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|