1
|
Zhang L. Effects of mixed biocrusts on soil nutrients and bacterial community structure: a case study from Hilly Loess Plateau, China. Sci Rep 2024; 14:21265. [PMID: 39261650 PMCID: PMC11391072 DOI: 10.1038/s41598-024-71927-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 09/02/2024] [Indexed: 09/13/2024] Open
Abstract
The ecological function of biological crusts in arid and semi-arid areas is of great importance. Bacteria, as a crucial microbial group in biological crusts, play a key role in the formation, nutrient cycling, and regulation of these crusts. However, the succession of biological crusts and the diversity of bacterial communities, along with key environmental factors in the Loess Plateau's hilly and gully areas, remain unclear. This study investigated soil bacterial abundance and diversity in bare soil (BS), alga-lichen mixed crust (MC), and alga-lichen mixed crust subsoil (MCS) using high-throughput sequencing methods. It explored the relationship between the bacterial community in biological crusts and key environmental factors. The results indicated that the Chao1, Shannon index, and phylogenetic diversity of bacteria significantly increased with the succession of biological crusts. There were notable differences in the community composition and structure of bacteria at different stages of crust development, with Rubrobacteria and Cyanobacteriia dominating in MCS. Effective phosphorus, available potassium, nitrogen, pH, and total organic carbon were identified as key environmental factors affecting soil bacterial communities. In summary, the succession of biological crusts alters soil physicochemical characteristics and creates different ecological niches for bacterial communities. Soil nutrients and pH play a crucial role in the selection of bacterial species and the shaping of bacterial communities in the Loess Plateau's hilly and gully areas.
Collapse
Affiliation(s)
- Lei Zhang
- Technology Lnnovation Center for Land Engineering and Human Seutlements, Xi'an Jiaotong University, Xi'an, 713599, China.
- Shaanxi Provincial Land Engineering Construction Group Co., Ltd., Xi'an, 710054, China.
| |
Collapse
|
2
|
Mohseni R, Mahdavi Sharif P, Behfar M, Modaresi MR, Shirzadi R, Mardani M, Jafari L, Jafari F, Nikfetrat Z, Hamidieh AA. Evaluation of safety and efficacy of allogeneic adipose tissue-derived mesenchymal stem cells in pediatric bronchiolitis obliterans syndrome (BoS) after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Stem Cell Res Ther 2023; 14:256. [PMID: 37726865 PMCID: PMC10510238 DOI: 10.1186/s13287-023-03498-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 09/13/2023] [Indexed: 09/21/2023] Open
Abstract
BACKGROUND Allo-HSCT is a definite approach for the management of a wide variety of lethal and debilitating malignant and non-malignant disorders. However, its two main complications, acute and chronic graft-versus-host disease (GVHD), exert significant morbidities and mortalities. BoS, as a manifestation of chronic lung GVHD, is a gruesome complication of allo-HSCT, and for those with steroid-refractory disease, no approved second-line therapies exist. Mesenchymal stem cells (MSCs) exert anti-inflammatory and growth-promoting effects, and their administration against a wide range of inflammatory and neurologic disorders, as well as GVHD, has been associated with promising outcomes. However, literature on the safety and effectiveness of MSC therapy for BoS and pediatric cGVHD is scarce. METHODS We designed a single-arm trial to administer adipose tissue (AT)-derived MSCs to pediatric patients with refractory BoS after allo-HSCT. AT-MSCs from obese, otherwise healthy donors were cultured in an ISO class 1 clean room and injected into the antecubital vein of eligible patients with a dose of 1 × 106/kg. The primary endpoints included a complete or partial response to therapy [in terms of increased forced expiratory volume in one second (FEV1) values and steroid dose reduction] and its safety profile. RESULTS Four eligible patients with a median age of 6.5 years were enrolled in the study. Steroid-induced osteoporosis and myopathy were present in three cases. A partial response was evident in three cases after a single injection of AT-MSCs. The treatment was safe and tolerable, and no treatment-related adverse events were noted. Two patients developed manageable COVID-19 infections one and 4 months after AT-MSC injection. After a median follow-up duration of 19 months, all cases are still alive and have had no indications for lung transplantation. CONCLUSIONS AT-MSCs could be safely administered to our pediatric cases with BoS post-allo-HSCT. Considering their advanced stage of disease, their sub-optimal functional capacity due to steroid-induced complications, and COVID-19 infection post-treatment, we believe that AT-MSC therapy can have possible efficacy in the management of pediatric BoS. The conduction of further studies with larger sample sizes and more frequent injections is prudent for further optimization of AT-MSC therapy against BoS. Trial registration Iranian Registry of Clinical Trials (IRCT), IRCT20201202049568N2. Registered 22 February 2021, https://en.irct.ir/trial/53143 .
Collapse
Affiliation(s)
- Rashin Mohseni
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center Hospital, Tehran University of Medical Sciences, 63 Qarib St., Keshavarz Blvd., Tehran, 14155-6559, 1419733161, Iran
| | - Pouya Mahdavi Sharif
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center Hospital, Tehran University of Medical Sciences, 63 Qarib St., Keshavarz Blvd., Tehran, 14155-6559, 1419733161, Iran
| | - Maryam Behfar
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center Hospital, Tehran University of Medical Sciences, 63 Qarib St., Keshavarz Blvd., Tehran, 14155-6559, 1419733161, Iran
| | - Mohammad Reza Modaresi
- Pediatric Respiratory and Sleep Medicine Research Center, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Rohola Shirzadi
- Pediatric Pulmonary Disease and Sleep Medicine Research Center, Pediatric Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahta Mardani
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center Hospital, Tehran University of Medical Sciences, 63 Qarib St., Keshavarz Blvd., Tehran, 14155-6559, 1419733161, Iran
| | - Leila Jafari
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center Hospital, Tehran University of Medical Sciences, 63 Qarib St., Keshavarz Blvd., Tehran, 14155-6559, 1419733161, Iran
| | - Fahimeh Jafari
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center Hospital, Tehran University of Medical Sciences, 63 Qarib St., Keshavarz Blvd., Tehran, 14155-6559, 1419733161, Iran
| | - Zeynab Nikfetrat
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center Hospital, Tehran University of Medical Sciences, 63 Qarib St., Keshavarz Blvd., Tehran, 14155-6559, 1419733161, Iran
| | - Amir Ali Hamidieh
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center Hospital, Tehran University of Medical Sciences, 63 Qarib St., Keshavarz Blvd., Tehran, 14155-6559, 1419733161, Iran.
| |
Collapse
|
3
|
Thanaskody K, Jusop AS, Tye GJ, Wan Kamarul Zaman WS, Dass SA, Nordin F. MSCs vs. iPSCs: Potential in therapeutic applications. Front Cell Dev Biol 2022; 10:1005926. [PMID: 36407112 PMCID: PMC9666898 DOI: 10.3389/fcell.2022.1005926] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 10/21/2022] [Indexed: 01/24/2023] Open
Abstract
Over the past 2 decades, mesenchymal stem cells (MSCs) have attracted a lot of interest as a unique therapeutic approach for a variety of diseases. MSCs are capable of self-renewal and multilineage differentiation capacity, immunomodulatory, and anti-inflammatory properties allowing it to play a role in regenerative medicine. Furthermore, MSCs are low in tumorigenicity and immune privileged, which permits the use of allogeneic MSCs for therapies that eliminate the need to collect MSCs directly from patients. Induced pluripotent stem cells (iPSCs) can be generated from adult cells through gene reprogramming with ectopic expression of specific pluripotency factors. Advancement in iPS technology avoids the destruction of embryos to make pluripotent cells, making it free of ethical concerns. iPSCs can self-renew and develop into a plethora of specialized cells making it a useful resource for regenerative medicine as they may be created from any human source. MSCs have also been used to treat individuals infected with the SARS-CoV-2 virus. MSCs have undergone more clinical trials than iPSCs due to high tumorigenicity, which can trigger oncogenic transformation. In this review, we discussed the overview of mesenchymal stem cells and induced pluripotent stem cells. We briefly present therapeutic approaches and COVID-19-related diseases using MSCs and iPSCs.
Collapse
Affiliation(s)
- Kalaiselvaan Thanaskody
- Centre for Tissue Engineering and Regenerative Medicine (CTERM), Faculty of Medicine, University Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Amirah Syamimi Jusop
- Centre for Tissue Engineering and Regenerative Medicine (CTERM), Faculty of Medicine, University Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Gee Jun Tye
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Gelugor, Malaysia
| | - Wan Safwani Wan Kamarul Zaman
- Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur, Malaysia,Centre for Innovation in Medical Engineering (CIME), Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Sylvia Annabel Dass
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Gelugor, Malaysia
| | - Fazlina Nordin
- Centre for Tissue Engineering and Regenerative Medicine (CTERM), Faculty of Medicine, University Kebangsaan Malaysia, Kuala Lumpur, Malaysia,*Correspondence: Fazlina Nordin,
| |
Collapse
|
4
|
Yuan W, Song HY, Xiong J, Jiang WL, Kang GJ, Huang J, Xie SP. Placenta‑derived mesenchymal stem cells ameliorate lipopolysaccharide‑induced inflammation in RAW264.7 cells and acute lung injury in rats. Mol Med Rep 2020; 22:1458-1466. [PMID: 32626979 PMCID: PMC7339743 DOI: 10.3892/mmr.2020.11231] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 04/14/2020] [Indexed: 02/06/2023] Open
Abstract
Acute lung injury (ALI) is a severe lung syndrome with high morbidity and mortality, due to its complex mechanism and lack of effective therapy. The use of placenta-derived mesenchymal stem cells (pMSCs) has provided novel insight into treatment options of ALI. The effects of pMSCs on lipopolysaccharide (LPS)-induced inflammation were studied using a co-culture protocol with LPS-stimulated RAW264.7 cells. An LPS-induced ALI Sprague-Dawley rat model was developed by intravenously injecting 7.5 mg/kg LPS, and intratracheal instillation of 1×105 pMSCs was performed after administration of LPS to investigate the therapeutic potential of these cells. pMSCs ameliorated LPS-induced ALI, as suggested by downregulated pro-inflammatory cytokine tumor necrosis factor-α and increased anti-inflammatory cytokine interleukin-10 in both cell and animal models. Moreover, the protein and leukocyte cells in bronchoalveolar lavage fluid decreased at a rapid rate after treatment with pMSCs. Histopathology demonstrated that pMSCs alleviated the infiltration of inflammatory cells, pulmonary hyperemia and hemorrhage, and interstitial edema. In addition, pMSC reduced the LPS-induced expression of C-X-C motif chemokine ligand 12 in RAW264.7 macrophages and in lung tissue of ALI rats. This demonstrated that pMSCs are therapeutically effective in LPS-induced ALI.
Collapse
Affiliation(s)
- Wen Yuan
- Department of Laboratory Medicine, Wuhan Children's Hospital, Wuhan Maternal and Child Healthcare Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430016, P.R. China
| | - Heng-Ya Song
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jie Xiong
- Department of Immunology, School of Basic Medical Science, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Wan-Li Jiang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Gan-Jun Kang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jie Huang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Song-Ping Xie
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
5
|
Gómez de Antonio D, Campo-Cañaveral de la Cruz JL, Zurita M, Santos M, González Lois C, Varela de Ugarte A, Vaquero J. Bone Marrow-derived Mesenchymal Stem Cells and Chronic Allograft Disease in a Bronchiolitis Obliterans Animal Model. Arch Bronconeumol 2020; 56:149-156. [PMID: 31296434 DOI: 10.1016/j.arbres.2019.05.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/22/2019] [Accepted: 05/23/2019] [Indexed: 10/26/2022]
Abstract
INTRODUCTION Bronchiolitis obliterans (BO) is the most common expression of chronic allograft dysfunction in lung transplantation. Moreover, BO represents the major cause of death in the long-term after this procedure. On the other hand, mesenchymal stem cells have been tested in animal models of BO aiming to interfere in its development. The aim of this experimental study is to explore the role of bone-marrow derived stem cells (BMSCs) as a preventive intervention of BO occurrence. MATERIALS AND METHODS This an experimental randomized study. A bronchiolitis obliterans animal model in rats was reproduced: heterotopical tracheal transplant model in lung parenchyma. Five of these animals were used as control group. After setting up the model, individuals were divided in 3 groups of treatment (n=15), in which BMSCs were administered in 3 different time points after the tracheal transplant (tracheal transplantation and BMSCs administration occurred the same day, group G0; after 7 days, group G7; after 14 days, group G14. In addition, within each group, BMSCs were administered through 3 different routes: endotracheally, endovascular and topically in the lung parenchyma). Animals were sacrificed at 21 days. Histology, fluorescence in situ hybridization and immunohistochemistry techniques were performed for identifying stem cells. RESULTS Compared to control group, animals receiving BMSCs showed large neovessels in a loose fibrous matrix. Group G7 showed less fibrosis (p<0.033) and edema (p<0.028). Moreover, G7 animals receiving stem cells endotracheally showed no fibrosis (p<0.008). Alveolar-like patches of tissue were observed among all groups (53.4%, 46.7% and 40% in G0, G7 and G14 respectively), consisting of cells expressing both stem and alveolar cells biomarkers. CONCLUSION BMSCs modify the course of bronchiolitis obliterans and differentiate into alveolar cells. Endotracheal administration of BMSCs 7 days after the heterotopical tracheal transplant might be considered an effective way to prevent BO in this animal model.
Collapse
Affiliation(s)
- David Gómez de Antonio
- Thoracic Surgery Department, Hospital Universitario Puerta de Hierro, Majadahonda, Spain.
| | | | - Mercedes Zurita
- Neuroscience Laboratory, Hospital Universitario Puerta de Hierro, Majadahonda, Spain
| | - Martin Santos
- Veterinary Department, Hospital Universitario Puerta de Hierro, Majadahonda, Spain
| | - Carmen González Lois
- Pathology Department, Hospital Universitario Puerta de Hierro, Majadahonda, Spain
| | | | - Jesús Vaquero
- Neuroscience Laboratory, Hospital Universitario Puerta de Hierro, Majadahonda, Spain
| |
Collapse
|
6
|
Postinfectious Bronchiolitis Obliterans in Children: Diagnostic Workup and Therapeutic Options: A Workshop Report. Can Respir J 2020; 2020:5852827. [PMID: 32076469 PMCID: PMC7013295 DOI: 10.1155/2020/5852827] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 11/29/2019] [Accepted: 12/27/2019] [Indexed: 12/23/2022] Open
Abstract
Bronchiolitis obliterans (BO) is a rare, chronic form of obstructive lung disease, often initiated with injury of the bronchiolar epithelium followed by an inflammatory response and progressive fibrosis of small airways resulting in nonuniform luminal obliteration or narrowing. The term BO comprises a group of diseases with different underlying etiologies, courses, and characteristics. Among the better recognized inciting stimuli leading to BO are airway pathogens such as adenovirus and mycoplasma, which, in a small percentage of infected children, will result in progressive fixed airflow obstruction, an entity referred to as postinfectious bronchiolitis obliterans (PIBO). The present knowledge on BO in general is reasonably well developed, in part because of the relatively high incidence in patients who have undergone lung transplantation or bone marrow transplant recipients who have had graft-versus-host disease in the posttransplant period. The cellular and molecular pathways involved in PIBO, while assumed to be similar, have not been adequately elucidated. Since 2016, an international consortium of experts with an interest in PIBO assembles on a regular basis in Geisenheim, Germany, to discuss key areas in PIBO which include diagnostic workup, treatment strategies, and research fields.
Collapse
|
7
|
Chen S, Zhao K, Lin R, Wang S, Fan Z, Huang F, Chen X, Nie D, Du X, Guo Z, Lin D, Xuan L, Xu N, Sun J, Peng Xiang A, Liu Q. The efficacy of mesenchymal stem cells in bronchiolitis obliterans syndrome after allogeneic HSCT: A multicenter prospective cohort study. EBioMedicine 2019; 49:213-222. [PMID: 31668569 PMCID: PMC6945279 DOI: 10.1016/j.ebiom.2019.09.039] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 09/21/2019] [Accepted: 09/22/2019] [Indexed: 12/19/2022] Open
Abstract
Background Bronchiolitis obliterans syndrome (BOS) after allo-HSCT is a devastating complication with limited therapeutic options. We aimed to assess the efficacy and safety of mesenchymal stem cells (MSCs) in BOS after allo-HSCT. Methods This multicenter prospective cohort study enrolled 81 allo-HSCT recipients whose BOS were diagnosed within 6 months. The choice of prednisone and azithromycin combined with or without MSCs was based on patient preferences (MSC n = 49, non-MSC n = 32). The primary endpoint was response rate at 3 months, defined as the proportion of patients achieving FEV1 improvement or steroid sparing. The trial was registered at ClinicalTrials.gov (NCT02543073). Findings Response rate was 35/49 patients (71%, 95% CI 59 to 84%) and 14/32 (44%, 27 to 61%) in MSC and non-MSC group, respectively (p = 0.013). The addition of MSCs was associated with a better difference for change in FEV1 rate of decline, compared to non-MSC group (53 mL/months, 2 to 103; p = 0.040). The 3-year overall survival post-diagnosis was 70.6% (55.9 to 85.3%) and 58.2% (36.1 to 78.5%) in MSC and non-MSC group, respectively (p = 0.21). Clinical improvement was accompanied by a significant increase of interleukin (IL)-10-producing CD5+B cells. There was no statistical difference in the rates of infections and leukemia relapse between the two groups. MSCs were well-tolerated with no serious adverse events. Interpretation MSCs offer an effective and safe therapeutic option for BOS after allo-HSCT. Our study strengthens evidence for clinical use of MSC therapy in BOS. These data also provide novel insight into potential biological mechanisms of MSC treatment and support further investigation in larger randomized controlled trials. Funding National Key R&D Program of China, National Natural Science Foundation of China, Health Collaborative Innovation Major Projects of Guangzhou City, Science and Technology Planning Project of Guangdong Province.
Collapse
Affiliation(s)
- Shan Chen
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ke Zhao
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ren Lin
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shunqing Wang
- Department of Hematology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zhiping Fan
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fen Huang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoyong Chen
- Center for Stem Cell Biology and Tissue Engineering, The Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Guangzhou, China
| | - Danian Nie
- Department of Hematology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xin Du
- Department of Hematology, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Ziwen Guo
- Department of Hematology, Zhongshan City People's Hospital, Zhongshan, China
| | - Dongjun Lin
- Department of Hematology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Li Xuan
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Na Xu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jing Sun
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, The Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Guangzhou, China; Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, China.
| | - Qifa Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
8
|
Ghazanfari T, Ghaffarpour S, Kariminia A, Salehi E, Hashemi SM, Ardestani SK, Gohari Moghadam K, Mirsharif ES, Dilmaghanian R, Fadaei A, Faghihzadeh S. Circulating mesenchymal stem cells in sulfur mustard-exposed patients with long-term pulmonary complications. Toxicol Lett 2019; 312:188-194. [PMID: 31095986 DOI: 10.1016/j.toxlet.2019.05.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 04/17/2019] [Accepted: 05/12/2019] [Indexed: 10/26/2022]
Abstract
Sulfur mustard (SM) is a toxic agent that causes acute and long-term pulmonary complications. Recent evidence has shown the impact of SM on mesenchymal stem cells (MSCs). These cells have a critical role in repairing the damaged tissues. In this study, we evaluated the mobilization of MSCs in SM-exposed patients with long-term pulmonary complications. Fifty-nine SM-injured patients with prolonged pulmonary complications and 20 healthy individuals were included. Patients were classified based on taking drugs, having comorbidities, and severity of respiratory consequence. MSCs with phenotype of CD45-CD44+CD29+CD105+ were evaluated in peripheral blood using flow cytometry. Circulating MSCs were lower in SM-exposed patients compared to the control group (0.93 vs. 2.72 respectively, P = 0.005). No significant difference was observed in the MSC count between patients taking corticosteroids or antibiotics and those patients not taking them. Comorbidities like liver and kidney diseases had changed the count of MSCs in SM-exposed subjects. In addition, the frequency of MSCs did not show any association with the severity of long-term pulmonary complications. In conclusion, SM-exposure causes a decline in the frequency of circulating MSCs in survivors. The lower number of the peripheral MSC population in SM-exposed patients was not affected by taking corticosteroids or antibiotics, but comorbidities are probably involved in MSC frequency. The decreases observed in the number of circulating MSCs was not associated with the severity of the pulmonary complications; however, further studies in mustard lung models are required to demonstrate the therapeutic or pathologic role of MSCs in SM injuries.
Collapse
Affiliation(s)
- Tooba Ghazanfari
- Immunoregulation Research Center, Shahed University, Tehran, Islamic Republic of Iran; Department of Immunology, Shahed University, 1471, North Karegar Street, Tehran, Islamic Republic of Iran.
| | - Sara Ghaffarpour
- Immunoregulation Research Center, Shahed University, Tehran, Islamic Republic of Iran
| | - Amina Kariminia
- British Columbia Children's Hospital Research Institute, Pediatrics Department, Faculty of, Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Eisa Salehi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medica, Sciences, Tehran, Islamic Republic of Iran
| | | | - Keivan Gohari Moghadam
- Internal Medicine Department, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | | | - Razieh Dilmaghanian
- Immunoregulation Research Center, Shahed University, Tehran, Islamic Republic of Iran
| | - Abbas Fadaei
- Department of Pulmonology and Intensive Care Medicine, Shahid Labbafinejad Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Soghrat Faghihzadeh
- Department of Biostatistics and Social Medicine, Zanjan University of Medical Sciences, Zanjan, Islamic Republic of Iran
| |
Collapse
|
9
|
Sher N, Ofir R. Placenta-Derived Adherent Stromal Cell Therapy for Hematopoietic Disorders: A Case Study of PLX-R18. Cell Transplant 2019; 27:140-150. [PMID: 29562777 PMCID: PMC6434483 DOI: 10.1177/0963689717727543] [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: 12/15/2022] Open
Abstract
The ephemeral placenta provides a noncontroversial source of young, healthy cells of both maternal and fetal origin from which cell therapy products can be manufactured. The 2 advantages of using live cells as therapeutic entities are: (a) in their environmental-responsive, multifactorial secretion profile and (b) in their activity as a “slow-release drug delivery system,” releasing secretions over a long time frame. A major difficulty in translating cell therapy to the clinic involves challenges of large-scale, robust manufacturing while maintaining product characteristics, identity, and efficacy. To address these concerns early on, Pluristem developed the PLacental eXpanded (PLX) platform, the first good manufacturing practice–approved, 3-dimensional bioreactor-based cell growth platform, to enable culture of mesenchymal-like adherent stromal cells harvested from the postpartum placenta. One of the products produced by Pluristem on this platform is PLX-R18, a product mainly comprising placental fetal cells, which is proven in vivo to alleviate radiation-induced lethality and to enhance hematopoietic cell counts after bone marrow (BM) failure. The identified mechanism of action of PLX-R18 cells is one of the cell-derived systemic pro-hematopoietic secretions, which upregulate endogenous secretions and subsequently rescue BM and peripheral blood cellularity, thereby boosting survival. PLX-R18 is therefore currently under study to treat both the hematopoietic syndrome of acute radiation (under the US Food and Drug Administration [FDA]’s Animal Rule) and the incomplete engraftment after BM transplantation (in a phase I study). In the future, they could potentially address additional hematological indications, such as aplastic anemia, myelodysplastic syndrome, primary graft failure, and acute or chronic graft versus host disease.
Collapse
|
10
|
Yamada Y, Brüstle K, Jungraithmayr W. T Helper Cell Subsets in Experimental Lung Allograft Rejection. J Surg Res 2018; 233:74-81. [PMID: 30502290 DOI: 10.1016/j.jss.2018.07.073] [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: 03/20/2018] [Revised: 06/10/2018] [Accepted: 07/23/2018] [Indexed: 01/05/2023]
Abstract
BACKGROUND Human lung transplantation has evolved to an established treatment for pulmonary diseases in their end stages; however, the long-term outcome is worse when compared to all other solid transplantable organs. The major reason for this unfavorable outcome is rejection, either in its acute or chronic form, the latter termed as chronic lung allograft dysfunction. METHODS A systematic review search was performed. RESULTS One of the most important immune cells responsible for rejection are T cells. Beside alloreactive CD8+ T cells, CD4+ T cells play a key role during the evolvement of allograft rejection. Certain subsets of these allograft CD4+ T cells have been identified which have been shown to exert either transplant-protective or transplant-injuring properties. These effects have been proven in various experimental models, mainly in rats and mice, and allowed for the gain of important insights into these proinflammatory and anti-inflammatory characteristics including their targetability: while the subsets Th1, Th17, Th22, and Tfh cells have been shown to act in a rather proinflammatory way, Tregs, Th2, and Th9 subsets exert anti-inflammatory effects. Chronic airway obstruction is mainly induced by IL17 as shown across models. CONCLUSIONS This review shall summarize and provide an overview of the current evidence about the role and effects of proinflammatory and anti-inflammatory CD4-+ T helper cell subsets during lung allograft rejection in experimental rodent models.
Collapse
Affiliation(s)
- Yoshito Yamada
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Karina Brüstle
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Wolfgang Jungraithmayr
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland; Department of Thoracic Surgery, Brandenburg Medical School, Neurupppin, Germany.
| |
Collapse
|
11
|
Yao Y, Zheng Z, Song Q. Mesenchymal stem cells: A double-edged sword in radiation-induced lung injury. Thorac Cancer 2017; 9:208-217. [PMID: 29235254 PMCID: PMC5792737 DOI: 10.1111/1759-7714.12573] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 11/10/2017] [Accepted: 11/10/2017] [Indexed: 01/06/2023] Open
Abstract
Radiation therapy is an important treatment modality for multiple thoracic malignancies. However, radiation‐induced lung injury (RILI), which is the term generally used to describe damage to the lungs caused by exposure to ionizing radiation, remains a critical issue affecting both tumor control and patient quality of life. Despite tremendous effort, there is no current consensus regarding the optimal treatment approach for RILI. Because of a number of functional advantages, including self‐proliferation, multi‐differentiation, injury foci chemotaxis, anti‐inflammation, and immunomodulation, mesenchymal stem cells (MSCs) have been a focus of research for many years. Accumulating evidence indicates the therapeutic potential of transplantation of MSCs derived from adipose tissue, umbilical cord blood, and bone marrow for inflammatory diseases, including RILI. However, reports have also shown that MSCs, including fibrocytes, lung hematopoietic progenitor cells, and ABCG2+ MSCs, actually enhance the progression of lung injuries. These contradictory results suggest that MSCs may have dual effects and that caution should be taken when using MSCs to treat RILI. In this review, we present and discuss recent evidence of the double‐edged function of MSCs and provide comments on the prospects of these findings.
Collapse
Affiliation(s)
- Yi Yao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhongliang Zheng
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Qibin Song
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
12
|
Zheng G, Qiu G, Ge M, He J, Huang L, Chen P, Wang W, Xu Q, Hu Y, Shu Q, Xu J. Human adipose-derived mesenchymal stem cells alleviate obliterative bronchiolitis in a murine model via IDO. Respir Res 2017; 18:119. [PMID: 28619045 PMCID: PMC5472885 DOI: 10.1186/s12931-017-0599-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 05/26/2017] [Indexed: 02/08/2023] Open
Abstract
Background Long-term survival of lung transplantation is hindered by the development of obliterative bronchiolitis (OB). Adipose-derived stem cells (ASCs) were documented to have more potent immunosuppressive ability than mesenchymal stem cells (MSCs) from bone marrow and placenta. The goal of our study is to evaluate the effect of repeated administration of ASCs on OB and the involvement of indoleamine 2,3-dioxygenase (IDO) mediating the protective effect of ASCs in a heterotopic tracheal transplantation (HTT) model. Methods For studies in vitro, ASCs were treated with interferon-γ (IFN-γ). For in vivo study, tracheas from BALB/c or C57BL/6 donors were transplanted into C57BL/6 recipients to create a HTT model. On days 0, 1, 3, 5, 8, 12, 15, 20 and 25 post-transplant, the allogeneic recipient mice were administered intravenously with phosphate buffered saline, 1 × 106 human ASCs, or 1 × 106 human ASCs plus 1-methyltryptophan (1-MT), an IDO inhibitor. On days 3, 7, 14 and 28, serum, trachea and spleen samples were harvested for analysis. Results ASCs homed to heterotopic tracheal grafts after infusion. Multiple doses of ASCs significantly increased tracheal IDO levels in allografts. There were significant increases in graft and serum IFN-γ levels in allografts compared with isografts. IFN-γ elevated IDO expression and activity in ASCs in vitro. ASCs alleviated OB in allografts as evidenced by reduced epithelial loss, epithelial apoptosis, and intraluminal obstruction. The effects of ASCs on OB were blocked by 1-MT. 1-MT also blocked the alterations in pro and anti-inflammatory cytokines as well as CD3+ T cell infiltration induced by ASCs. ASCs induced not only splenic levels of CD4+CD25+Foxp3+ regulatory T cells (Treg) but also IL-10 and TGF-β-producing Treg. Furthermore, IDO inhibition abolished the changes of splenic Treg induced by ASCs. In addition, Treg reduction by cyclophosphamide treatment did not alter the effects of ASCs on tracheal IDO expression in allografts confirming Treg induction is downstream of IDO. Conclusions Repeated doses of ASCs are capable of ameliorating OB. ASCs act at least in part via elevating IDO expression. ASCs promote the generation of Treg and suppress T cell infiltration via an IDO-dependent mechanism.
Collapse
Affiliation(s)
- Guoping Zheng
- Shaoxing Second Hospital, 123 Yanan Road, Shaoxing, Zhejiang, 312000, China
| | - Guanguan Qiu
- Shaoxing Second Hospital, 123 Yanan Road, Shaoxing, Zhejiang, 312000, China
| | - Menghua Ge
- Shaoxing Second Hospital, 123 Yanan Road, Shaoxing, Zhejiang, 312000, China
| | - Jianping He
- Shaoxing Second Hospital, 123 Yanan Road, Shaoxing, Zhejiang, 312000, China
| | - Lanfang Huang
- Shaoxing Second Hospital, 123 Yanan Road, Shaoxing, Zhejiang, 312000, China
| | - Ping Chen
- Shaoxing Second Hospital, 123 Yanan Road, Shaoxing, Zhejiang, 312000, China
| | - Wei Wang
- Shaoxing Second Hospital, 123 Yanan Road, Shaoxing, Zhejiang, 312000, China
| | - Qi Xu
- The Children's Hospital of Zhejiang University School of Medicine, 3333 Binsheng Road, Hangzhou, Zhejiang, 310051, China
| | - Yaoqin Hu
- The Children's Hospital of Zhejiang University School of Medicine, 3333 Binsheng Road, Hangzhou, Zhejiang, 310051, China
| | - Qiang Shu
- The Children's Hospital of Zhejiang University School of Medicine, 3333 Binsheng Road, Hangzhou, Zhejiang, 310051, China.
| | - Jianguo Xu
- Shaoxing Second Hospital, 123 Yanan Road, Shaoxing, Zhejiang, 312000, China. .,The First Affiliated Hospital of Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, Zhejiang, 310003, China.
| |
Collapse
|
13
|
Tu ZL, Zhou ZY, Xu HC, Cao JL, Ye P, Wang LM, Lv W, Hu J. LTB4 and montelukast in transplantation-related bronchiolitis obliterans in rats. J Cardiothorac Surg 2017; 12:43. [PMID: 28545478 PMCID: PMC5445521 DOI: 10.1186/s13019-017-0605-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 05/11/2017] [Indexed: 02/07/2023] Open
Abstract
Background Lung transplantation is the only effective treatment for end-stage lung diseases. Bronchiolitis obliterans, which is known as non-infectious chronic lung allograft dysfunction (CLAD) in the new classification, is the greatest threat to long-term survival after lung transplantation. This study investigated the role of leukotriene B4 (LTB4) and montelukast in transplantation-related bronchiolitis obliterans and discussed the pathophysiological significance of LTB4 in chronic rejection. Methods Rats were randomly divided into an experimental group (montelukast), a positive control group (dexamethasone), and a blank control group (normal saline solution; NS). Each piece of trachea removed from a F344 rat was transplanted into a Lewis rat through a 5-mm incision at the episternum by subcutaneous embedding. The recipients were treated with gastric lavage with 3 mg/kg · d montelukast suspension, 1 mg/kg · d dexamethasone, and 1 mL/kg · d NS, respectively, in each group. On Day 28, peripheral blood was drawn to measure the white blood cell counts and plasma LTB4 levels. The donor specimens were stained by H-E and Masson, and their organizational structure and extent of fibrosis were visually assessed. The measurement data were compared using one-way analysis of variance, and the categorical data were compared using the chi-square test. A P value of less than 0.05 was considered to indicate statistical significance. Results The white blood cell counts of the montelukast, dexamethasone, and NS groups were (16.0 ± 4.2) × 109/L, (19.5 ± 11.6) × 109/L, and (25.8 ± 3.6) × 109/L; no statistical significance was found (P = 0.101). The concentrations of LTB4 were 2230 ± 592 pg/mL, 1961 ± 922 pg/mL, and 3764 ± 1169 pg/mL, and statistical significance was found between the NS group and each of the others (P = 0.009). The percentages of tracheal occlusion were 73.6% ± 13.8%, 23.4% ± 3.2%, and 89.9% ± 11.3%, and statistical significance was found among the three groups (P = 0.000). Conclusions The study established a model to simulate bronchiolitis obliterans after clinical lung transplantation. Oral administration of montelukast reduced plasma LTB4 levels in rats and played a preventive role against tracheal fibrosis after transplantation. This suggests that LTB4 may be involved in bronchiolitis obliterans after pulmonary transplantation. This study indicates a new direction for research into the prevention and treatment of bronchiolitis obliterans after lung transplantation.
Collapse
Affiliation(s)
- Zheng-Liang Tu
- Department of Thoracic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, NO. 79 Qingchun Road, Hangzhou, 310003, China
| | - Zhen-Yu Zhou
- Department of Thoracic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, NO. 79 Qingchun Road, Hangzhou, 310003, China
| | - Hai-Chao Xu
- Department of Thoracic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, NO. 79 Qingchun Road, Hangzhou, 310003, China
| | - Jin-Lin Cao
- Department of Thoracic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, NO. 79 Qingchun Road, Hangzhou, 310003, China
| | - Peng Ye
- Department of Thoracic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, NO. 79 Qingchun Road, Hangzhou, 310003, China
| | - Lu-Ming Wang
- Department of Thoracic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, NO. 79 Qingchun Road, Hangzhou, 310003, China
| | - Wang Lv
- Department of Thoracic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, NO. 79 Qingchun Road, Hangzhou, 310003, China
| | - Jian Hu
- Department of Thoracic Surgery, the First Affiliated Hospital, College of Medicine, Zhejiang University, NO. 79 Qingchun Road, Hangzhou, 310003, China.
| |
Collapse
|
14
|
Lorenzi W, Gonçalves FDC, Schneider N, Silva ÉF, Visioli F, Paz AH, Saueressig MG. Repeated systemic administration of adipose tissue-derived mesenchymal stem cells prevents tracheal obliteration in a murine model of bronchiolitis obliterans. Biotechnol Lett 2017; 39:1269-1277. [PMID: 28528427 DOI: 10.1007/s10529-017-2355-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 05/04/2017] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To investigate the effect of adipose tissue-derived mesenchymal stem cell (ASC) administered either systemically or locally in a murine model of bronchiolitis obliterans. RESULTS When compared to controls, systemic treatment with 106 ASCs on D0 and a second dose on D7 significantly prevented tracheal obliteration 28 days after heterotopic tracheal transplantation (median of 94 vs. 16%; P < 0.01). A single dose tended towards less stenosis than controls, but did not reach statistical significance (28 vs. 94%; P = 0.054). On the contrary, repeated local injection was incapable of preventing tracheal obliteration when compared to a single injection or controls (37 vs. 71 vs. 87%). Two intravenous doses also tended to be better than two local injections (16 vs. 37%; P = 0.058), and were better than a single local dose (16 vs. 71%; P < 0.01). CONCLUSION A second dose of ASC, given systemically after 7 days, reduces luminal obliteration in a heterotopic tracheal transplantation model in mice, suggesting that ASC can be used to prevent obliterative bronchiolitis after lung transplantation.
Collapse
Affiliation(s)
- William Lorenzi
- Thoracic Surgery Service, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, Santa Cecília, Porto Alegre, RS, CEP 90035-903, Brazil.
| | - Fabiany Da Costa Gonçalves
- Experimental Research Center, Hospital de Clínicas de Porto Alegre, Ramiro Barcelos 2350, Porto Alegre, RS, CEP 90035-903, Brazil
| | - Natália Schneider
- Experimental Research Center, Hospital de Clínicas de Porto Alegre, Ramiro Barcelos 2350, Porto Alegre, RS, CEP 90035-903, Brazil
| | - Éverton Franco Silva
- Thoracic Surgery Service, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, Santa Cecília, Porto Alegre, RS, CEP 90035-903, Brazil
| | - Fernanda Visioli
- Pathology Department, School of Odontology, Universidade Federal do Rio Grande do Sul, Ramiro Barcelos 2492, Porto Alegre, RS, CEP 90035-007, Brazil
| | - Ana Helena Paz
- Experimental Research Center, Hospital de Clínicas de Porto Alegre, Ramiro Barcelos 2350, Porto Alegre, RS, CEP 90035-903, Brazil
| | - Mauricio Guidi Saueressig
- Thoracic Surgery Service, Hospital de Clínicas de Porto Alegre, Rua Ramiro Barcelos, 2350, Santa Cecília, Porto Alegre, RS, CEP 90035-903, Brazil
| |
Collapse
|
15
|
Abumaree MH, Abomaray FM, Alshabibi MA, AlAskar AS, Kalionis B. Immunomodulatory properties of human placental mesenchymal stem/stromal cells. Placenta 2017; 59:87-95. [PMID: 28411943 DOI: 10.1016/j.placenta.2017.04.003] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 03/14/2017] [Accepted: 04/06/2017] [Indexed: 02/09/2023]
Abstract
Mesenchymal stem/stromal cells (MSCs) are isolated from various fetal and adult tissues such as bone marrow, adipose tissue, cord blood and placenta. Placental MSCs (pMSCs), the main focus of this review, are relatively new MSC types that are not as intensively studied compared with bone marrow-derived MSCs (BMMSCs). MSCs modulate the immune functions of important immune cells involved in alloantigen recognition and elimination, including antigen presenting cells (APCs), T cells, B cells and natural killer (NK) cells. Clinical trials, both completed and underway, employ MSCs to treat various human immunological diseases, such as multiple sclerosis (MS) and type 1 diabetes. However, the mechanisms that mediate the immunosuppressive effects of pMSCs are still largely unknown, and the safety of pMSC use in clinical settings needs further confirmation. Here, we review the current knowledge of the immunosuppressive properties of placental MSCs.
Collapse
Affiliation(s)
- M H Abumaree
- Stem Cells and Regenerative Medicine Department, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Minstry of National Guard Health Affairs, P.O. Box 22490, Riyadh 11426, Mail Code 1515, Saudi Arabia; College of Science and Health Professions, King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Minstry of National Guard Health Affairs, P.O. Box 3660, Riyadh 11481, Mail Code 3124, Saudi Arabia.
| | - F M Abomaray
- Department of Clinical Science, Intervention and Technology, Division of Obstetrics and Gynecology, Karolinska Institutet, 14186 Stockholm, Sweden; Center for Hematology and Regenerative Medicine, Karolinska Institutet, 14186 Stockholm, Sweden
| | - M A Alshabibi
- National Center for Stem Cell Technology, Life Sciences and Environment Research Institute, King Abdulaziz City for Science and Technology, P.O Box 6086, Riyadh 11442, Saudi Arabia
| | - A S AlAskar
- Stem Cells and Regenerative Medicine Department, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Minstry of National Guard Health Affairs, P.O. Box 22490, Riyadh 11426, Mail Code 1515, Saudi Arabia
| | - B Kalionis
- Department of Maternal-Fetal Medicine Pregnancy Research Centre, University of Melbourne Department of Obstetrics and Gynaecology, Royal Women's Hospital, Parkville, Victoria, 3052, Australia
| |
Collapse
|
16
|
Mordant P, Nakajima D, Kalaf R, Iskender I, Maahs L, Behrens P, Coutinho R, Iyer RK, Davies JE, Cypel M, Liu M, Waddell TK, Keshavjee S. Mesenchymal stem cell treatment is associated with decreased perfusate concentration of interleukin-8 during ex vivo perfusion of donor lungs after 18-hour preservation. J Heart Lung Transplant 2016; 35:1245-1254. [DOI: 10.1016/j.healun.2016.04.017] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 03/28/2016] [Accepted: 04/25/2016] [Indexed: 01/16/2023] Open
|
17
|
Gao L, Zhang Y, Hu B, Liu J, Kong P, Lou S, Su Y, Yang T, Li H, Liu Y, Zhang C, Gao L, Zhu L, Wen Q, Wang P, Chen X, Zhong J, Zhang X. Phase II Multicenter, Randomized, Double-Blind Controlled Study of Efficacy and Safety of Umbilical Cord-Derived Mesenchymal Stromal Cells in the Prophylaxis of Chronic Graft-Versus-Host Disease After HLA-Haploidentical Stem-Cell Transplantation. J Clin Oncol 2016; 34:2843-50. [PMID: 27400949 DOI: 10.1200/jco.2015.65.3642] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
PURPOSE Although mesenchymal stromal cells (MSCs) possess immunomodulatory properties and exhibit promising efficacy against chronic graft-versus-host disease (cGVHD), little is known about the efficacy of MSCs in the prophylaxis of cGVHD after HLA-haploidentical hematopoietic stem-cell transplantation (HLA-haplo HSCT). PATIENTS AND METHODS In this multicenter, double-blind, randomized controlled trial, we investigated the incidence and severity of cGVHD among patients, and the changes in T, B, and natural killer (NK) cells after the repeated infusion of MSCs. RESULTS The 2-year cumulative incidence of cGVHD in the MSCs group was 27.4% (95% CI, 16.2% to 38.6%), compared with 49.0% (95% CI, 36.5% to 61.5%) in the non-MSCs control group (P = .021). Seven patients in the non-MSCs control group had severe lung cGVHD, but no patients in the MSCs group developed typical lung cGVHD (P = .047). After the MSC infusions, increasing memory B lymphocytes and regulatory T cells, as well as the ratio of type 1 T helper to type 2 T helper cells, were observed, whereas the number of NK cells decreased. CONCLUSION Our findings suggest that the repeated infusion of MSCs might inhibit cGVHD symptoms in patients after HLA-haplo HSCT, accompanied by changes in the numbers and subtypes of T, B, and NK cells, leading to the acquisition of immune tolerance.
Collapse
Affiliation(s)
- Lei Gao
- Lei Gao, Yanqi Zhang, Jia Liu, Peiyan Kong, Yao Liu, Li Gao, Cheng Zhang, Lidan Zhu, Qin Wen, Ping Wang, Xinghua Chen, Jiangfan Zhong, and Xi Zhang, Third Military Medical University, Chongqing; Baoyang Hu, Chinese Academy of Sciences, Beijing; Shifeng Lou, Second Affiliated Hospital of Chongqing Medical University, Chongqing; Yi Su, General Hospital of Chengdu Military Region of People's Liberation Army, Chengdu; Tonghua Yang, Yunnan Provincial People's Hospital; Huimin Li, Affiliated Hospital of Kunming Medical College, Kunming, China; and Jiangfan Zhong, University of Southern California, Los Angeles, CA
| | - Yanqi Zhang
- Lei Gao, Yanqi Zhang, Jia Liu, Peiyan Kong, Yao Liu, Li Gao, Cheng Zhang, Lidan Zhu, Qin Wen, Ping Wang, Xinghua Chen, Jiangfan Zhong, and Xi Zhang, Third Military Medical University, Chongqing; Baoyang Hu, Chinese Academy of Sciences, Beijing; Shifeng Lou, Second Affiliated Hospital of Chongqing Medical University, Chongqing; Yi Su, General Hospital of Chengdu Military Region of People's Liberation Army, Chengdu; Tonghua Yang, Yunnan Provincial People's Hospital; Huimin Li, Affiliated Hospital of Kunming Medical College, Kunming, China; and Jiangfan Zhong, University of Southern California, Los Angeles, CA
| | - Baoyang Hu
- Lei Gao, Yanqi Zhang, Jia Liu, Peiyan Kong, Yao Liu, Li Gao, Cheng Zhang, Lidan Zhu, Qin Wen, Ping Wang, Xinghua Chen, Jiangfan Zhong, and Xi Zhang, Third Military Medical University, Chongqing; Baoyang Hu, Chinese Academy of Sciences, Beijing; Shifeng Lou, Second Affiliated Hospital of Chongqing Medical University, Chongqing; Yi Su, General Hospital of Chengdu Military Region of People's Liberation Army, Chengdu; Tonghua Yang, Yunnan Provincial People's Hospital; Huimin Li, Affiliated Hospital of Kunming Medical College, Kunming, China; and Jiangfan Zhong, University of Southern California, Los Angeles, CA
| | - Jia Liu
- Lei Gao, Yanqi Zhang, Jia Liu, Peiyan Kong, Yao Liu, Li Gao, Cheng Zhang, Lidan Zhu, Qin Wen, Ping Wang, Xinghua Chen, Jiangfan Zhong, and Xi Zhang, Third Military Medical University, Chongqing; Baoyang Hu, Chinese Academy of Sciences, Beijing; Shifeng Lou, Second Affiliated Hospital of Chongqing Medical University, Chongqing; Yi Su, General Hospital of Chengdu Military Region of People's Liberation Army, Chengdu; Tonghua Yang, Yunnan Provincial People's Hospital; Huimin Li, Affiliated Hospital of Kunming Medical College, Kunming, China; and Jiangfan Zhong, University of Southern California, Los Angeles, CA
| | - Peiyan Kong
- Lei Gao, Yanqi Zhang, Jia Liu, Peiyan Kong, Yao Liu, Li Gao, Cheng Zhang, Lidan Zhu, Qin Wen, Ping Wang, Xinghua Chen, Jiangfan Zhong, and Xi Zhang, Third Military Medical University, Chongqing; Baoyang Hu, Chinese Academy of Sciences, Beijing; Shifeng Lou, Second Affiliated Hospital of Chongqing Medical University, Chongqing; Yi Su, General Hospital of Chengdu Military Region of People's Liberation Army, Chengdu; Tonghua Yang, Yunnan Provincial People's Hospital; Huimin Li, Affiliated Hospital of Kunming Medical College, Kunming, China; and Jiangfan Zhong, University of Southern California, Los Angeles, CA
| | - Shifeng Lou
- Lei Gao, Yanqi Zhang, Jia Liu, Peiyan Kong, Yao Liu, Li Gao, Cheng Zhang, Lidan Zhu, Qin Wen, Ping Wang, Xinghua Chen, Jiangfan Zhong, and Xi Zhang, Third Military Medical University, Chongqing; Baoyang Hu, Chinese Academy of Sciences, Beijing; Shifeng Lou, Second Affiliated Hospital of Chongqing Medical University, Chongqing; Yi Su, General Hospital of Chengdu Military Region of People's Liberation Army, Chengdu; Tonghua Yang, Yunnan Provincial People's Hospital; Huimin Li, Affiliated Hospital of Kunming Medical College, Kunming, China; and Jiangfan Zhong, University of Southern California, Los Angeles, CA
| | - Yi Su
- Lei Gao, Yanqi Zhang, Jia Liu, Peiyan Kong, Yao Liu, Li Gao, Cheng Zhang, Lidan Zhu, Qin Wen, Ping Wang, Xinghua Chen, Jiangfan Zhong, and Xi Zhang, Third Military Medical University, Chongqing; Baoyang Hu, Chinese Academy of Sciences, Beijing; Shifeng Lou, Second Affiliated Hospital of Chongqing Medical University, Chongqing; Yi Su, General Hospital of Chengdu Military Region of People's Liberation Army, Chengdu; Tonghua Yang, Yunnan Provincial People's Hospital; Huimin Li, Affiliated Hospital of Kunming Medical College, Kunming, China; and Jiangfan Zhong, University of Southern California, Los Angeles, CA
| | - Tonghua Yang
- Lei Gao, Yanqi Zhang, Jia Liu, Peiyan Kong, Yao Liu, Li Gao, Cheng Zhang, Lidan Zhu, Qin Wen, Ping Wang, Xinghua Chen, Jiangfan Zhong, and Xi Zhang, Third Military Medical University, Chongqing; Baoyang Hu, Chinese Academy of Sciences, Beijing; Shifeng Lou, Second Affiliated Hospital of Chongqing Medical University, Chongqing; Yi Su, General Hospital of Chengdu Military Region of People's Liberation Army, Chengdu; Tonghua Yang, Yunnan Provincial People's Hospital; Huimin Li, Affiliated Hospital of Kunming Medical College, Kunming, China; and Jiangfan Zhong, University of Southern California, Los Angeles, CA
| | - Huimin Li
- Lei Gao, Yanqi Zhang, Jia Liu, Peiyan Kong, Yao Liu, Li Gao, Cheng Zhang, Lidan Zhu, Qin Wen, Ping Wang, Xinghua Chen, Jiangfan Zhong, and Xi Zhang, Third Military Medical University, Chongqing; Baoyang Hu, Chinese Academy of Sciences, Beijing; Shifeng Lou, Second Affiliated Hospital of Chongqing Medical University, Chongqing; Yi Su, General Hospital of Chengdu Military Region of People's Liberation Army, Chengdu; Tonghua Yang, Yunnan Provincial People's Hospital; Huimin Li, Affiliated Hospital of Kunming Medical College, Kunming, China; and Jiangfan Zhong, University of Southern California, Los Angeles, CA
| | - Yao Liu
- Lei Gao, Yanqi Zhang, Jia Liu, Peiyan Kong, Yao Liu, Li Gao, Cheng Zhang, Lidan Zhu, Qin Wen, Ping Wang, Xinghua Chen, Jiangfan Zhong, and Xi Zhang, Third Military Medical University, Chongqing; Baoyang Hu, Chinese Academy of Sciences, Beijing; Shifeng Lou, Second Affiliated Hospital of Chongqing Medical University, Chongqing; Yi Su, General Hospital of Chengdu Military Region of People's Liberation Army, Chengdu; Tonghua Yang, Yunnan Provincial People's Hospital; Huimin Li, Affiliated Hospital of Kunming Medical College, Kunming, China; and Jiangfan Zhong, University of Southern California, Los Angeles, CA
| | - Cheng Zhang
- Lei Gao, Yanqi Zhang, Jia Liu, Peiyan Kong, Yao Liu, Li Gao, Cheng Zhang, Lidan Zhu, Qin Wen, Ping Wang, Xinghua Chen, Jiangfan Zhong, and Xi Zhang, Third Military Medical University, Chongqing; Baoyang Hu, Chinese Academy of Sciences, Beijing; Shifeng Lou, Second Affiliated Hospital of Chongqing Medical University, Chongqing; Yi Su, General Hospital of Chengdu Military Region of People's Liberation Army, Chengdu; Tonghua Yang, Yunnan Provincial People's Hospital; Huimin Li, Affiliated Hospital of Kunming Medical College, Kunming, China; and Jiangfan Zhong, University of Southern California, Los Angeles, CA
| | - Li Gao
- Lei Gao, Yanqi Zhang, Jia Liu, Peiyan Kong, Yao Liu, Li Gao, Cheng Zhang, Lidan Zhu, Qin Wen, Ping Wang, Xinghua Chen, Jiangfan Zhong, and Xi Zhang, Third Military Medical University, Chongqing; Baoyang Hu, Chinese Academy of Sciences, Beijing; Shifeng Lou, Second Affiliated Hospital of Chongqing Medical University, Chongqing; Yi Su, General Hospital of Chengdu Military Region of People's Liberation Army, Chengdu; Tonghua Yang, Yunnan Provincial People's Hospital; Huimin Li, Affiliated Hospital of Kunming Medical College, Kunming, China; and Jiangfan Zhong, University of Southern California, Los Angeles, CA
| | - Lidan Zhu
- Lei Gao, Yanqi Zhang, Jia Liu, Peiyan Kong, Yao Liu, Li Gao, Cheng Zhang, Lidan Zhu, Qin Wen, Ping Wang, Xinghua Chen, Jiangfan Zhong, and Xi Zhang, Third Military Medical University, Chongqing; Baoyang Hu, Chinese Academy of Sciences, Beijing; Shifeng Lou, Second Affiliated Hospital of Chongqing Medical University, Chongqing; Yi Su, General Hospital of Chengdu Military Region of People's Liberation Army, Chengdu; Tonghua Yang, Yunnan Provincial People's Hospital; Huimin Li, Affiliated Hospital of Kunming Medical College, Kunming, China; and Jiangfan Zhong, University of Southern California, Los Angeles, CA
| | - Qin Wen
- Lei Gao, Yanqi Zhang, Jia Liu, Peiyan Kong, Yao Liu, Li Gao, Cheng Zhang, Lidan Zhu, Qin Wen, Ping Wang, Xinghua Chen, Jiangfan Zhong, and Xi Zhang, Third Military Medical University, Chongqing; Baoyang Hu, Chinese Academy of Sciences, Beijing; Shifeng Lou, Second Affiliated Hospital of Chongqing Medical University, Chongqing; Yi Su, General Hospital of Chengdu Military Region of People's Liberation Army, Chengdu; Tonghua Yang, Yunnan Provincial People's Hospital; Huimin Li, Affiliated Hospital of Kunming Medical College, Kunming, China; and Jiangfan Zhong, University of Southern California, Los Angeles, CA
| | - Ping Wang
- Lei Gao, Yanqi Zhang, Jia Liu, Peiyan Kong, Yao Liu, Li Gao, Cheng Zhang, Lidan Zhu, Qin Wen, Ping Wang, Xinghua Chen, Jiangfan Zhong, and Xi Zhang, Third Military Medical University, Chongqing; Baoyang Hu, Chinese Academy of Sciences, Beijing; Shifeng Lou, Second Affiliated Hospital of Chongqing Medical University, Chongqing; Yi Su, General Hospital of Chengdu Military Region of People's Liberation Army, Chengdu; Tonghua Yang, Yunnan Provincial People's Hospital; Huimin Li, Affiliated Hospital of Kunming Medical College, Kunming, China; and Jiangfan Zhong, University of Southern California, Los Angeles, CA
| | - Xinghua Chen
- Lei Gao, Yanqi Zhang, Jia Liu, Peiyan Kong, Yao Liu, Li Gao, Cheng Zhang, Lidan Zhu, Qin Wen, Ping Wang, Xinghua Chen, Jiangfan Zhong, and Xi Zhang, Third Military Medical University, Chongqing; Baoyang Hu, Chinese Academy of Sciences, Beijing; Shifeng Lou, Second Affiliated Hospital of Chongqing Medical University, Chongqing; Yi Su, General Hospital of Chengdu Military Region of People's Liberation Army, Chengdu; Tonghua Yang, Yunnan Provincial People's Hospital; Huimin Li, Affiliated Hospital of Kunming Medical College, Kunming, China; and Jiangfan Zhong, University of Southern California, Los Angeles, CA
| | - Jiangfan Zhong
- Lei Gao, Yanqi Zhang, Jia Liu, Peiyan Kong, Yao Liu, Li Gao, Cheng Zhang, Lidan Zhu, Qin Wen, Ping Wang, Xinghua Chen, Jiangfan Zhong, and Xi Zhang, Third Military Medical University, Chongqing; Baoyang Hu, Chinese Academy of Sciences, Beijing; Shifeng Lou, Second Affiliated Hospital of Chongqing Medical University, Chongqing; Yi Su, General Hospital of Chengdu Military Region of People's Liberation Army, Chengdu; Tonghua Yang, Yunnan Provincial People's Hospital; Huimin Li, Affiliated Hospital of Kunming Medical College, Kunming, China; and Jiangfan Zhong, University of Southern California, Los Angeles, CA
| | - Xi Zhang
- Lei Gao, Yanqi Zhang, Jia Liu, Peiyan Kong, Yao Liu, Li Gao, Cheng Zhang, Lidan Zhu, Qin Wen, Ping Wang, Xinghua Chen, Jiangfan Zhong, and Xi Zhang, Third Military Medical University, Chongqing; Baoyang Hu, Chinese Academy of Sciences, Beijing; Shifeng Lou, Second Affiliated Hospital of Chongqing Medical University, Chongqing; Yi Su, General Hospital of Chengdu Military Region of People's Liberation Army, Chengdu; Tonghua Yang, Yunnan Provincial People's Hospital; Huimin Li, Affiliated Hospital of Kunming Medical College, Kunming, China; and Jiangfan Zhong, University of Southern California, Los Angeles, CA.
| |
Collapse
|
18
|
Tooi M, Komaki M, Morioka C, Honda I, Iwasaki K, Yokoyama N, Ayame H, Izumi Y, Morita I. Placenta Mesenchymal Stem Cell Derived Exosomes Confer Plasticity on Fibroblasts. J Cell Biochem 2016; 117:1658-70. [PMID: 26640165 DOI: 10.1002/jcb.25459] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 12/04/2015] [Indexed: 12/22/2022]
Abstract
Mesenchymal stem cell (MSC)-conditioned medium (MSC-CM) has been reported to enhance wound healing. Exosomes contain nucleic acids, proteins, and lipids, and function as an intercellular communication vehicle for mediating some paracrine effects. However, the function of MSC-derived exosomes (MSC-exo) remains elusive. In this study, we isolated human placenta MSC (PlaMSC)-derived exosomes (PlaMSC-exo) and examined their function in vitro. PlaMSCs were isolated from human term placenta using enzymatic digestion. PlaMSC-exo were prepared from the conditioned medium of PlaMSC (PlaMSC-CM) by ultracentrifugation. The expression of stemness-related genes, such as OCT4 and NANOG, in normal adult human dermal fibroblasts (NHDF) after incubation with PlaMSC-exo was measured by real-time reverse transcriptase PCR analysis (real-time PCR). The effect of PlaMSC-exo on OCT4 transcription activity was assessed using Oct4-EGFP reporter mice-derived dermal fibroblasts. The stimulating effects of PlaMSC-exo on osteoblastic and adipocyte-differentiation of NHDF were evaluated by alkaline phosphatase (ALP), and Alizarin red S- and oil red O-staining, respectively. The expression of osteoblast- and adipocyte-related genes was also assessed by real-time PCR. The treatment of NHDF with PlaMSC-exo significantly upregulated OCT4 and NANOG mRNA expression. PlaMSC-exo also enhanced OCT4 transcription. The NHDF treated with PlaMSC-exo exhibited osteoblastic and adipocyte-differentiation in osteogenic and adipogenic induction media. PlaMSC-exo increase the expression of OCT4 and NANOG mRNA in fibroblasts. As a result, PlaMSC-exo influence the differentiation competence of fibroblasts to both osteoblastic and adipocyte-differentiation. It shows a new feature of MSCs and the possibility of clinical application of MSC-exo. J. Cell. Biochem. 117: 1658-1670, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Masayuki Tooi
- Department of Periodontology, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Motohiro Komaki
- Department of Nanomedicine (DNP), Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Chikako Morioka
- Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Izumi Honda
- Department of Comprehensive Reproductive Medicine, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Kengo Iwasaki
- Department of Nanomedicine (DNP), Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Naoki Yokoyama
- Life Science Laboratory, Research and Development Center, Dai Nippon Printing Co., Ltd., 1-1-1 Kaga-cho, Shinjuku-ku, Tokyo, 162-8001, Japan
| | - Hirohito Ayame
- Life Science Laboratory, Research and Development Center, Dai Nippon Printing Co., Ltd., 1-1-1 Kaga-cho, Shinjuku-ku, Tokyo, 162-8001, Japan
| | - Yuichi Izumi
- Department of Periodontology, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Ikuo Morita
- Department of Cellular Physiological Chemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| |
Collapse
|
19
|
Zhao Y, Gillen JR, Meher AK, Burns JA, Kron IL, Lau CL. Rapamycin prevents bronchiolitis obliterans through increasing infiltration of regulatory B cells in a murine tracheal transplantation model. J Thorac Cardiovasc Surg 2016; 151:487-96.e3. [PMID: 26481278 PMCID: PMC4728002 DOI: 10.1016/j.jtcvs.2015.08.116] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 07/06/2015] [Accepted: 08/11/2015] [Indexed: 02/05/2023]
Abstract
OBJECTIVE B lymphocytes are generally considered to be activators of the immune response; however, recent findings have shown that a subtype of B lymphocytes, regulatory B lymphocytes, play a role in attenuating the immune response. Bronchiolitis obliterans remains the major limitation to modern-day lung transplantation. The role of regulatory B lymphocytes in bronchiolitis obliterans has not been elucidated. We hypothesized that regulatory B lymphocytes play a role in the attenuation of bronchiolitis obliterans. METHODS We performed a standard heterotopic tracheal transplant model. Tracheas from Balb/c mice were transplanted into C57BL/6 recipients. Rapamycin treatment and dimethyl sulfoxide control groups were each treated for the first 14 days after the transplant. Tracheas were collected on days 7, 14, and 28 post-transplantation. Luminal obliteration was evaluated by hematoxylin-eosin staining and Picrosirius red staining. Immune cell infiltration and characteristics, and secretion of interleukin-10 and transforming growth factor-β1 were accessed by immunohistochemistry. Cytokines and transforming growth factor-β1 were measured using the Luminex assay (Bio-Rad, Hercules, Calif). RESULTS The results revealed that intraperitoneal injection of rapamycin for 14 days after tracheal transplantation significantly reduced luminal obliteration on day 28 when compared with the dimethyl sulfoxide control group (97.78% ± 3.63% vs 3.02% ± 2.14%, P < .001). Rapamycin treatment markedly induced regulatory B lymphocytes (B220(+)IgM(+)IgG(-)IL-10(+)TGF-β1(+)) cells when compared with dimethyl sulfoxide controls. Rapamycin treatment inhibited interleukin-1β, 6, 13, and 17 on days 7 and 14. Rapamycin also greatly increased interleukin-10 and transforming growth factor-β1 production in B cells and regulatory T lymphocytes infiltration on day 28. CONCLUSIONS Mammalian target of rapamycin inhibition decreases the development of bronchiolitis obliterans via inhibition of proinflammatory cytokines and increasing regulatory B lymphocytes cell infiltration, which subsequently produces anti-inflammatory cytokines and upregulates regulatory T lymphocyte cells.
Collapse
Affiliation(s)
- Yunge Zhao
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, University of Virginia, Charlottesville, Va
| | - Jacob R Gillen
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, University of Virginia, Charlottesville, Va
| | - Akshaya K Meher
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, University of Virginia, Charlottesville, Va
| | - Jordan A Burns
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, University of Virginia, Charlottesville, Va
| | - Irving L Kron
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, University of Virginia, Charlottesville, Va
| | - Christine L Lau
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, University of Virginia, Charlottesville, Va.
| |
Collapse
|
20
|
Placenta as a Source of Stem Cells for Regenerative Medicine. CURRENT PATHOBIOLOGY REPORTS 2015. [DOI: 10.1007/s40139-015-0070-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
21
|
Raza K, Larsen T, Samaratunga N, Price AP, Meyer C, Matson A, Ehrhardt MJ, Fogas S, Tolar J, Hertz MI, Panoskaltsis-Mortari A. MSC therapy attenuates obliterative bronchiolitis after murine bone marrow transplant. PLoS One 2014; 9:e109034. [PMID: 25272285 PMCID: PMC4182803 DOI: 10.1371/journal.pone.0109034] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 09/03/2014] [Indexed: 12/18/2022] Open
Abstract
RATIONALE Obliterative bronchiolitis (OB) is a significant cause of morbidity and mortality after lung transplant and hematopoietic cell transplant. Mesenchymal stromal cells (MSCs) have been shown to possess immunomodulatory properties in chronic inflammatory disease. OBJECTIVE Administration of MSCs was evaluated for the ability to ameliorate OB in mice using our established allogeneic bone marrow transplant (BMT) model. METHODS Mice were lethally conditioned and received allogeneic bone marrow without (BM) or with spleen cells (BMS), as a source of OB-causing T-cells. Cell therapy was started at 2 weeks post-transplant, or delayed to 4 weeks when mice developed airway injury, defined as increased airway resistance measured by pulmonary function test (PFT). BM-derived MSC or control cells [mouse pulmonary vein endothelial cells (PVECs) or lung fibroblasts (LFs)] were administered. Route of administration [intratracheally (IT) and IV] and frequency (every 1, 2 or 3 weeks) were compared. Mice were evaluated at 3 months post-BMT. MEASUREMENTS AND MAIN RESULTS No ectopic tissue formation was identified in any mice. When compared to BMS mice receiving control cells or no cells, those receiving MSCs showed improved resistance, compliance and inspiratory capacity. Interim PFT analysis showed no difference in route of administration. Improvements in PFTs were found regardless of dose frequency; but once per week worked best even when administration began late. Mice given MSC also had decreased peribronchiolar inflammation, lower levels of hydroxyproline (collagen) and higher frequencies of macrophages staining for the alternatively activated macrophage (AAM) marker CD206. CONCLUSIONS These results warrant study of MSCs as a potential management option for OB in lung transplant and BMT recipients.
Collapse
Affiliation(s)
- Kashif Raza
- Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Trevor Larsen
- Breck High School, Edina, Minnesota, United States of America
| | | | - Andrew P Price
- Pediatric Blood and Bone Marrow Transplant Program, University of Minnesota Cancer Center, Minneapolis, Minnesota, United States of America
| | - Carolyn Meyer
- Pediatric Blood and Bone Marrow Transplant Program, University of Minnesota Cancer Center, Minneapolis, Minnesota, United States of America
| | - Amy Matson
- Pediatric Blood and Bone Marrow Transplant Program, University of Minnesota Cancer Center, Minneapolis, Minnesota, United States of America
| | - Michael J Ehrhardt
- Pediatric Blood and Bone Marrow Transplant Program, University of Minnesota Cancer Center, Minneapolis, Minnesota, United States of America
| | - Samuel Fogas
- Pediatric Blood and Bone Marrow Transplant Program, University of Minnesota Cancer Center, Minneapolis, Minnesota, United States of America
| | - Jakub Tolar
- Pediatric Blood and Bone Marrow Transplant Program, University of Minnesota Cancer Center, Minneapolis, Minnesota, United States of America
| | - Marshall I Hertz
- Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Angela Panoskaltsis-Mortari
- Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Minnesota, Minneapolis, Minnesota, United States of America; Pediatric Blood and Bone Marrow Transplant Program, University of Minnesota Cancer Center, Minneapolis, Minnesota, United States of America
| |
Collapse
|
22
|
Gracon ASA, Wilkes DS. Lung transplantation: chronic allograft dysfunction and establishing immune tolerance. Hum Immunol 2014; 75:887-94. [PMID: 24979671 PMCID: PMC4357397 DOI: 10.1016/j.humimm.2014.06.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 06/19/2014] [Accepted: 06/19/2014] [Indexed: 10/25/2022]
Abstract
Despite significant medical advances since the advent of lung transplantation, improvements in long-term survival have been largely unrealized. Chronic lung allograft dysfunction, in particular obliterative bronchiolitis, is the primary limiting factor. The predominant etiology of obliterative bronchiolitis involves the recipient's innate and adaptive immune response to the transplanted allograft. Current therapeutic strategies have failed to provide a definitive treatment paradigm to improve long-term outcomes. Inducing immune tolerance is an emerging therapeutic strategy that abrogates allograft rejection, avoids immunosuppression, and improves long-term graft function. The aim of this review is to discuss the key immunologic components of obliterative bronchiolitis, describe the state of establishing immune tolerance in transplantation, and highlight those strategies being evaluated in lung transplantation.
Collapse
Affiliation(s)
- Adam S A Gracon
- Department of Surgery and Center for Immunobiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - David S Wilkes
- Departments of Medicine, Microbiology and Immunology, Center for Immunobiology, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|