451
|
Arango-Rodriguez ML, Ezquer F, Ezquer M, Conget P. Could cancer and infection be adverse effects of mesenchymal stromal cell therapy? World J Stem Cells 2015; 7:408-417. [PMID: 25815124 PMCID: PMC4369496 DOI: 10.4252/wjsc.v7.i2.408] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Revised: 10/01/2014] [Accepted: 11/10/2014] [Indexed: 02/06/2023] Open
Abstract
Multipotent mesenchymal stromal cells [also referred to as mesenchymal stem cells (MSCs)] are a heterogeneous subset of stromal cells. They can be isolated from bone marrow and many other types of tissue. MSCs are currently being tested for therapeutic purposes (i.e., improving hematopoietic stem cell engraftment, managing inflammatory diseases and regenerating damaged organs). Their tropism for tumors and inflamed sites and their context-dependent potential for producing trophic and immunomodulatory factors raises the question as to whether MSCs promote cancer and/or infection. This article reviews the effect of MSCs on tumor establishment, growth and metastasis and also susceptibility to infection and its progression. Data published to date shows a paradoxical effect regarding MSCs, which seems to depend on isolation and expansion, cells source and dose and the route and timing of administration. Cancer and infection may thus be adverse or therapeutic effects arising form MSC administration.
Collapse
|
452
|
Liu GY, Liu Y, Lu Y, Qin YR, Di GH, Lei YH, Liu HX, Li YQ, Wu C, Hu XW, Duan HF. Short-term memory of danger signals or environmental stimuli in mesenchymal stem cells: implications for therapeutic potential. Cell Mol Immunol 2015; 13:369-78. [PMID: 25942600 DOI: 10.1038/cmi.2015.11] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 01/21/2015] [Accepted: 01/21/2015] [Indexed: 12/27/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) possess some characteristics of immune cells, including a pro-inflammatory phenotype, an immunosuppressive phenotype, antibacterial properties and the expression of Toll-like receptor proteins. Here we show that, similar to immune cells, MSCs retain information from danger signals or environmental stimuli for a period of time. When treated with the pro-inflammatory factors lipopolysaccharide (LPS) or tumor necrosis factor-α (TNF-α), MSCs display increased expression of IL-6, IL-8 and MCP-1. Following re-plating and several rounds of cell division in the absence of stimulating factors, the expression of IL-6, IL-8 and MCP-1 remained higher than in untreated cells for over 7 days. A spike in cytokine secretion occurred when cells were exposed to a second round of stimulation. We primed MSCs with LPS and LPS-primed MSCs had better therapeutic efficacy at promoting skin flap survival in a diabetic rat model than did unprimed MSCs. Finally, we found that several microRNAs, including miR146a, miR150 and miR155, along with the modification of DNA by 5-hydroxymethylcytosine (5hmC), mediate the MSC response to LPS and TNF-α stimulation. Collectively, our data suggest that MSCs have a short-term memory of environmental signals, which may impact their therapeutic potential.
Collapse
Affiliation(s)
- Guang-Yang Liu
- Key Laboratory of Systems Bioengineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
| | - Yang Liu
- Beijing Institute of Radiation Medicine (BIRM), Beijing, China
| | - Ying Lu
- 307 Hospital of the Chinese People's Liberation Army, Beijing, China
| | - Ya-Ru Qin
- Beijing Institute of Radiation Medicine (BIRM), Beijing, China
| | - Guo-Hu Di
- Beijing Institute of Radiation Medicine (BIRM), Beijing, China
| | - Yong-Hong Lei
- 301 Hospital of the Chinese People's Liberation Army, Beijing, China
| | - Hu-Xian Liu
- 301 Hospital of the Chinese People's Liberation Army, Beijing, China
| | - Yan-Qi Li
- Key Laboratory of Systems Bioengineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
| | - Chutse Wu
- Key Laboratory of Systems Bioengineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, China.,Beijing Institute of Radiation Medicine (BIRM), Beijing, China
| | - Xian-Wen Hu
- Beijing Institute of Biotechnology (BIB), Beijing, China
| | - Hai-Feng Duan
- Beijing Institute of Radiation Medicine (BIRM), Beijing, China
| |
Collapse
|
453
|
Lu H, Poirier C, Cook T, Traktuev DO, Merfeld-Clauss S, Lease B, Petrache I, March KL, Bogatcheva NV. Conditioned media from adipose stromal cells limit lipopolysaccharide-induced lung injury, endothelial hyperpermeability and apoptosis. J Transl Med 2015; 13:67. [PMID: 25889857 PMCID: PMC4358867 DOI: 10.1186/s12967-015-0422-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 01/28/2015] [Indexed: 01/08/2023] Open
Abstract
Background Acute Respiratory Distress Syndrome (ARDS) is a condition that contributes to morbidity and mortality of critically ill patients. We investigated whether factors secreted by adipose stromal cells (ASC) into conditioned media (ASC-CM) will effectively decrease lung injury in the model of lipopolysaccharide (LPS)-induced ARDS. Methods To assess the effect of ASC-CM on ARDS indices, intravenous delivery of ASC and ASC-CM to C57Bl/6 mice was carried out 4 h after LPS oropharyngeal aspiration; Evans Blue Dye (EBD) was injected intravenously 1 h prior to animal sacrifice (48 h post-LPS). Lungs were either fixed for histopathology, or used to extract bronchoalveolar lavage fluid (BALF) or EBD. To assess the effect of ASC-CM on endothelial barrier function and apoptosis, human pulmonary artery endothelial cells were treated with ASC-CM for 48-72 h. Results ASC-CM markedly reduced LPS-induced histopathologic changes of lung, protein extravasation into BALF, and suppressed the secretion of proinflammatory cytokines TNFα and IL6. White Blood Cells (WBC) from BALF of LPS-challenged mice receiving ASC-CM had decreased reactive oxygen species (ROS) generation compared to WBC from LPS-challenged mice receiving control media injection. Treatment of pulmonary endothelial monolayers with ASC-CM significantly suppressed H2O2-induced leakage of FITC dextran and changes in transendothelial resistance, as well as gap formation in endothelial monolayer. ASC-CM exposure reduced the percentage of endothelial cells expressing ICAM-1, and suppressed TNFα-induced expression of E-selectin and cleavage of caspase-3. ASC-CM reduced the endothelial level of pro-apoptotic protein Bim, but did not affect the level of Bcl-2, Bad, or Bad phosphorylation. Conclusions Factors secreted by ASC efficiently reduce ARDS indices, endothelial barrier hyperpermeability, and activation of pro-inflammatory and pro-apoptotic pathways in endothelium.
Collapse
Affiliation(s)
- Hongyan Lu
- Division of Cardiology, Indiana University, Indianapolis, IN, USA. .,Indiana Center for Vascular Biology and Medicine and VC-CAST Signature Center, Indianapolis, IN, USA. .,Roudebush Veterans Affairs Medical Center, Indiana University, Indianapolis, IN, USA.
| | - Christophe Poirier
- Division of Pulmonary and Critical Care Medicine, Indiana University, Indianapolis, IN, USA.
| | - Todd Cook
- Division of Cardiology, Indiana University, Indianapolis, IN, USA. .,Indiana Center for Vascular Biology and Medicine and VC-CAST Signature Center, Indianapolis, IN, USA.
| | - Dmitry O Traktuev
- Division of Cardiology, Indiana University, Indianapolis, IN, USA. .,Indiana Center for Vascular Biology and Medicine and VC-CAST Signature Center, Indianapolis, IN, USA. .,Roudebush Veterans Affairs Medical Center, Indiana University, Indianapolis, IN, USA.
| | - Stephanie Merfeld-Clauss
- Division of Cardiology, Indiana University, Indianapolis, IN, USA. .,Indiana Center for Vascular Biology and Medicine and VC-CAST Signature Center, Indianapolis, IN, USA. .,Roudebush Veterans Affairs Medical Center, Indiana University, Indianapolis, IN, USA.
| | - Benjamin Lease
- Indiana Center for Vascular Biology and Medicine and VC-CAST Signature Center, Indianapolis, IN, USA.
| | - Irina Petrache
- Indiana Center for Vascular Biology and Medicine and VC-CAST Signature Center, Indianapolis, IN, USA. .,Roudebush Veterans Affairs Medical Center, Indiana University, Indianapolis, IN, USA. .,Division of Pulmonary and Critical Care Medicine, Indiana University, Indianapolis, IN, USA.
| | - Keith L March
- Division of Cardiology, Indiana University, Indianapolis, IN, USA. .,Indiana Center for Vascular Biology and Medicine and VC-CAST Signature Center, Indianapolis, IN, USA. .,Roudebush Veterans Affairs Medical Center, Indiana University, Indianapolis, IN, USA.
| | - Natalia V Bogatcheva
- Division of Cardiology, Indiana University, Indianapolis, IN, USA. .,Indiana Center for Vascular Biology and Medicine and VC-CAST Signature Center, Indianapolis, IN, USA. .,Roudebush Veterans Affairs Medical Center, Indiana University, Indianapolis, IN, USA.
| |
Collapse
|
454
|
Zhao YD, Huang X, Yi F, Dai Z, Qian Z, Tiruppathi C, Tran K, Zhao YY. Endothelial FoxM1 mediates bone marrow progenitor cell-induced vascular repair and resolution of inflammation following inflammatory lung injury. Stem Cells 2015; 32:1855-64. [PMID: 24578354 DOI: 10.1002/stem.1690] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 01/25/2014] [Accepted: 01/30/2014] [Indexed: 01/08/2023]
Abstract
Adult stem cell treatment is a potential novel therapeutic approach for acute respiratory distress syndrome. Given the extremely low rate of cell engraftment, it is believed that these cells exert their beneficial effects via paracrine mechanisms. However, the endogenous mediator(s) in the pulmonary vasculature remains unclear. Using the mouse model with endothelial cell (EC)-restricted disruption of FoxM1 (FoxM1 CKO), here we show that endothelial expression of the reparative transcriptional factor FoxM1 is required for the protective effects of bone marrow progenitor cells (BMPC) against LPS-induced inflammatory lung injury and mortality. BMPC treatment resulted in rapid induction of FoxM1 expression in wild type (WT) but not FoxM1 CKO lungs. BMPC-induced inhibition of lung vascular injury, resolution of lung inflammation, and survival, as seen in WT mice, were abrogated in FoxM1 CKO mice following LPS challenge. Mechanistically, BMPC treatment failed to induce lung EC proliferation in FoxM1 CKO mice, which was associated with impaired expression of FoxM1 target genes essential for cell cycle progression. We also observed that BMPC treatment enhanced endothelial barrier function in WT but not in FoxM1-deficient EC monolayers. Restoration of β-catenin expression in FoxM1-deficient ECs normalized endothelial barrier enhancement in response to BMPC treatment. These data demonstrate the requisite role of endothelial FoxM1 in the mechanism of BMPC-induced vascular repair to restore vascular integrity and accelerate resolution of inflammation, thereby promoting survival following inflammatory lung injury.
Collapse
Affiliation(s)
- Yidan D Zhao
- Department of Pharmacology, The University of Illinois College of Medicine, Chicago, Illinois, USA; Center for Lung and Vascular Biology, The University of Illinois College of Medicine, Chicago, Illinois, USA
| | | | | | | | | | | | | | | |
Collapse
|
455
|
Andrews KL, Houdek MT, Kiemele LJ. Wound management of chronic diabetic foot ulcers: from the basics to regenerative medicine. Prosthet Orthot Int 2015; 39:29-39. [PMID: 25614499 DOI: 10.1177/0309364614534296] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND Hospital-based studies have shown that mortality rates in individuals with diabetic foot ulcers are about twice those observed in individuals with diabetes without foot ulcers. OBJECTIVE To assess the etiology and management of chronic diabetic foot ulcers. STUDY DESIGN Literature review. METHODS Systematic review of the literature discussing management of diabetic foot ulcers. Since there were only a few randomized controlled trials on this topic, articles were selected to attempt to be comprehensive rather than a formal assessment of study quality. RESULTS Chronic nonhealing foot ulcers occur in approximately 15% of patients with diabetes. Many factors contribute to impaired diabetic wound healing. Risk factors include peripheral neuropathy, peripheral arterial disease, limited joint mobility, foot deformities, abnormal foot pressures, minor trauma, a history of ulceration or amputation, and impaired visual acuity. With the current treatment for nonhealing diabetic foot ulcers, a significant number of patients require amputation. CONCLUSION Diabetic foot ulcers are optimally managed by a multidisciplinary integrated team. Offloading and preventative management are important. Dressings play an adjunctive role. There is a critical need to develop novel treatments to improve healing of diabetic foot ulcers. The goal is to have wounds heal and remain healed. CLINICAL RELEVANCE Diabetic neuropathy and peripheral arterial disease are major factors involved in a diabetic foot ulcer. Despite current treatment modalities for nonhealing diabetic foot ulcers, there are a significant number of patients who require amputations. No known therapy will be effective without concomitant management of ischemia, infection, and adequate offloading.
Collapse
Affiliation(s)
- Karen L Andrews
- Vascular Ulcer/Wound Healing Clinic, Gonda Vascular Center, Mayo Clinic, Rochester, MN, USA Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | - Matthew T Houdek
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Lester J Kiemele
- Vascular Ulcer/Wound Healing Clinic, Gonda Vascular Center, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
456
|
Wilson JG, Liu KD, Zhuo H, Caballero L, McMillan M, Fang X, Cosgrove K, Vojnik R, Calfee CS, Lee JW, Rogers AJ, Levitt J, Wiener-Kronish J, Bajwa EK, Leavitt A, McKenna D, Thompson BT, Matthay MA. Mesenchymal stem (stromal) cells for treatment of ARDS: a phase 1 clinical trial. THE LANCET. RESPIRATORY MEDICINE 2015; 3:24-32. [PMID: 25529339 PMCID: PMC4297579 DOI: 10.1016/s2213-2600(14)70291-7] [Citation(s) in RCA: 565] [Impact Index Per Article: 62.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND No effective pharmacotherapy for acute respiratory distress syndrome (ARDS) exists, and mortality remains high. Preclinical studies support the efficacy of mesenchymal stem (stromal) cells (MSCs) in the treatment of lung injury. We aimed to test the safety of a single dose of allogeneic bone marrow-derived MSCs in patients with moderate-to-severe ARDS. METHODS The STem cells for ARDS Treatment (START) trial was a multicentre, open-label, dose-escalation, phase 1 clinical trial. Patients were enrolled in the intensive care units at University of California, San Francisco, CA, USA, Stanford University, Stanford, CA, USA, and Massachusetts General Hospital, Boston, MA, USA, between July 8, 2013, and Jan 13, 2014. Patients were included if they had moderate-to-severe ARDS as defined by the acute onset of the need for positive pressure ventilation by an endotracheal or tracheal tube, a PaO2:FiO2 less than 200 mm Hg with at least 8 cm H2O positive end-expiratory airway pressure (PEEP), and bilateral infiltrates consistent with pulmonary oedema on frontal chest radiograph. The first three patients were treated with low dose MSCs (1 million cells/kg predicted bodyweight [PBW]), the next three patients received intermediate dose MSCs (5 million cells/kg PBW), and the final three patients received high dose MSCs (10 million cells/kg PBW). Primary outcomes included the incidence of prespecified infusion-associated events and serious adverse events. The trial is registered with ClinicalTrials.gov, number NCT01775774. FINDINGS No prespecified infusion-associated events or treatment-related adverse events were reported in any of the nine patients. Serious adverse events were subsequently noted in three patients during the weeks after the infusion: one patient died on study day 9, one patient died on study day 31, and one patient was discovered to have multiple embolic infarcts of the spleen, kidneys, and brain that were age-indeterminate, but thought to have occurred before the MSC infusion based on MRI results. None of these severe adverse events were thought to be MSC-related. INTERPRETATION A single intravenous infusion of allogeneic, bone marrow-derived human MSCs was well tolerated in nine patients with moderate to severe ARDS. Based on this phase 1 experience, we have proceeded to phase 2 testing of MSCs for moderate to severe ARDS with a primary focus on safety and secondary outcomes including respiratory, systemic, and biological endpoints. FUNDING The National Heart, Lung, and Blood Institute.
Collapse
Affiliation(s)
- Jennifer G. Wilson
- Departments of Emergency Medicine and Anesthesia, University of California, San Francisco
| | - Kathleen D. Liu
- Departments of Nephrology and Anesthesia, University of California, San Francisco
| | - Hanjing Zhuo
- Cardiovascular Research Institute, University of California, San Francisco
| | - Lizette Caballero
- Clinical Laboratories, Blood and Marrow Transplant Laboratory, University of California, San Francisco
| | - Melanie McMillan
- Clinical Laboratories, Blood and Marrow Transplant Laboratory, University of California, San Francisco
| | - Xiaohui Fang
- Cardiovascular Research Institute, University of California, San Francisco
| | - Katherine Cosgrove
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital
| | - Rosemary Vojnik
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University
| | - Carolyn S. Calfee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco
| | - Jae-Woo Lee
- Department of Anesthesia, University of California, San Francisco
| | - Angela J. Rogers
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University
| | - Joseph Levitt
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University
| | - Jeanine Wiener-Kronish
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard University
| | - Ednan K. Bajwa
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital
| | - Andrew Leavitt
- Department of Laboratory Medicine, University of California, San Francisco
| | - David McKenna
- Division of Transfusion Medicine, Department of Laboratory Medicine and Pathology, University of Minnesota
| | - B. Taylor Thompson
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital
| | - Michael A. Matthay
- Departments of Medicine and Anesthesia and the Cardiovascular Research Institute, University of California, San Francisco
| |
Collapse
|
457
|
Cell-based therapy for acute organ injury: preclinical evidence and ongoing clinical trials using mesenchymal stem cells. Anesthesiology 2014; 121:1099-121. [PMID: 25211170 DOI: 10.1097/aln.0000000000000446] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Critically ill patients often suffer from multiple organ failures involving lung, kidney, liver, or brain. Genomic, proteomic, and metabolomic approaches highlight common injury mechanisms leading to acute organ failure. This underlines the need to focus on therapeutic strategies affecting multiple injury pathways. The use of adult stem cells such as mesenchymal stem or stromal cells (MSC) may represent a promising new therapeutic approach as increasing evidence shows that MSC can exert protective effects following injury through the release of promitotic, antiapoptotic, antiinflammatory, and immunomodulatory soluble factors. Furthermore, they can mitigate metabolomic and oxidative stress imbalance. In this work, the authors review the biological capabilities of MSC and the results of clinical trials using MSC as therapy in acute organ injuries. Although preliminary results are encouraging, more studies concerning safety and efficacy of MSC therapy are needed to determine their optimal clinical use. (ANESTHESIOLOGY 2014; 121:1099-121).
Collapse
|
458
|
Gotts JE, Matthay MA. Endogenous and exogenous cell-based pathways for recovery from acute respiratory distress syndrome. Clin Chest Med 2014; 35:797-809. [PMID: 25453426 PMCID: PMC4254691 DOI: 10.1016/j.ccm.2014.08.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Regenerative medicine has entered a rapid phase of discovery, and much has been learned in recent years about the lung's response to injury. This article first summarizes the cellular and molecular mechanisms that damage the alveolar-capillary barrier, producing acute respiratory distress syndrome (ARDS). The latest understanding of endogenous repair processes is discussed, highlighting the diversity of lung epithelial progenitor cell populations and their regulation in health and disease. Finally, the past, present, and future of exogenous cell-based therapies for ARDS is reviewed.
Collapse
Affiliation(s)
- Jeffrey E Gotts
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, 505 Parnassus Avenue, San Francisco, CA 94143-0624, USA
| | - Michael A Matthay
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, 505 Parnassus Avenue, San Francisco, CA 94143-0624, USA.
| |
Collapse
|
459
|
Vandermeulen M, Grégoire C, Briquet A, Lechanteur C, Beguin Y, Detry O. Rationale for the potential use of mesenchymal stromal cells in liver transplantation. World J Gastroenterol 2014; 20:16418-16432. [PMID: 25469010 PMCID: PMC4248185 DOI: 10.3748/wjg.v20.i44.16418] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 08/28/2014] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are multipotent and self-renewing cells that reside essentially in the bone marrow as a non-hematopoietic cell population, but may also be isolated from the connective tissues of most organs. MSCs represent a heterogeneous population of adult, fibroblast-like cells characterized by their ability to differentiate into tissues of mesodermal lineages including adipocytes, chondrocytes and osteocytes. For several years now, MSCs have been evaluated for their in vivo and in vitro immunomodulatory and ‘tissue reconstruction’ properties, which could make them interesting in various clinical settings, and particularly in organ transplantation. This paper aims to review current knowledge on the properties of MSCs and their use in pre-clinical and clinical studies in solid organ transplantation, and particularly in the field of liver transplantation. The first available clinical data seem to show that MSCs are safe to use, at least in the medium-term, but more time is needed to evaluate the potential adverse effects of long-term use. Many issues must be resolved on the correct use of MSCs. Intensive in vitro and pre-clinical research are the keys to a better understanding of the way that MSCs act, and to eventually lead to clinical success.
Collapse
|
460
|
Zhu YG, Feng XM, Abbott J, Fang XH, Hao Q, Monsel A, Qu JM, Matthay MA, Lee JW. Human mesenchymal stem cell microvesicles for treatment of Escherichia coli endotoxin-induced acute lung injury in mice. Stem Cells 2014; 32:116-25. [PMID: 23939814 DOI: 10.1002/stem.1504] [Citation(s) in RCA: 492] [Impact Index Per Article: 49.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 07/27/2013] [Indexed: 12/13/2022]
Abstract
We previously found that human mesenchymal stem cells (MSC) or its conditioned medium restored lung protein permeability and reduced alveolar inflammation following Escherichia coli endotoxin-induced acute lung injury (ALI) in an ex vivo perfused human lung in part through the secretion of soluble factors such as keratinocyte growth factor (KGF). Recently, MSC were found to release microvesicles (MVs) that were biologically active because of the presence of mRNA or miRNA with reparative properties. MVs are circular fragments of membrane released from the endosomal compartment as exosomes or shed from the surface membranes. These studies were designed to determine if MVs released by human bone marrow derived MSCs would be effective in restoring lung protein permeability and reducing inflammation in E. coli endotoxin-induced ALI in C57BL/6 mice. The intratracheal instillation of MVs improved several indices of ALI at 48 hours. Compared to endotoxin-injured mice, MVs reduced extravascular lung water by 43% and reduced total protein levels in the bronchoalveolar lavage (BAL) fluid by 35%, demonstrating a reduction in pulmonary edema and lung protein permeability. MVs also reduced the influx of neutrophils and macrophage inflammatory protein-2 levels in the BAL fluid by 73% and 49%, respectively, demonstrating a reduction in inflammation. KGF siRNA-pretreatment of MSC partially eliminated the therapeutic effects of MVs released by MSCs, suggesting that KGF protein expression was important for the underlying mechanism. In summary, human MSC-derived MVs were therapeutically effective following E. coli endotoxin-induced ALI in mice in part through the expression of KGF mRNA in the injured alveolus.
Collapse
Affiliation(s)
- Ying-Gang Zhu
- Department of Pulmonary Medicine, Huadong Hospital, Fudan University, Shanghai, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
461
|
Biedermann A, Kriebel K, Kreikemeyer B, Lang H. Interactions of anaerobic bacteria with dental stem cells: an in vitro study. PLoS One 2014; 9:e110616. [PMID: 25369260 PMCID: PMC4219685 DOI: 10.1371/journal.pone.0110616] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Accepted: 09/15/2014] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND In patients with periodontitis, it is highly likely that local (progenitor) cells encounter pathogenic bacteria. The purpose of this in vitro study was to elucidate how human dental follicle stem cells (hDFSC) react towards a direct challenge with anaerobic periodontal pathogens under their natural oxygen-free atmosphere. HDFSC were compared to human bone marrow mesenchymal stem cells (hBMSC) and differentiated primary human gingival fibroblasts (hGiF), as well as permanent gingival carcinoma cells (Ca9-22). METHODOLOGY/PRINCIPAL FINDINGS The different cell types were investigated in a co-culture system with Porphyromonas gingivalis (P. gingivalis) and Fusobacterium nucleatum (F. nucleatum). The viability of the cells and pathogens under anaerobic conditions, as well as interactions in terms of adherence and internalization, were examined. Additionally, the release of pro-inflammatory interleukin-8 (IL-8) and anti-inflammatory interleukin-10 (IL-10) was quantified via enzyme-linked immunosorbent assay. The bacteria adhered less efficiently to hDFSC compared to Ca9-22 (P. gingivalis: 0.18% adherence to hDFSC; 3.1% adherence to Ca9-22). Similar results were observed for host cell internalization (F. nucleatum: 0.002% internalization into hDFSC; 0.09% internalization into Ca9-22). Statistically significantly less IL-8 was secreted from hDFSC after stimulation with F. nucleatum and P. gingivalis in comparison with hGiF (F. nucleatum: 2080.0 pg/ml--hGiF; 19.7 pg/ml--hDFSC). The IL-10 response of the differentiated cells was found to be low in relation to their pro-inflammatory IL-8 response. CONCLUSIONS/SIGNIFICANCE The results indicate that dental stem cells are less prone to interactions with pathogenic bacteria than differentiated cells in an anaerobic environment. Moreover, during bacterial challenge, the stem cell immune response seems to be more towards an anti-inflammatory reaction. For a potential future therapeutic use of hDFSC, these findings support the idea of a save application.
Collapse
Affiliation(s)
- Anne Biedermann
- Department of Operative Dentistry and Periodontology, University of Rostock, Rostock, Germany
| | - Katja Kriebel
- Department of Operative Dentistry and Periodontology, University of Rostock, Rostock, Germany
| | - Bernd Kreikemeyer
- Institute of Med. Microbiology, Virology and Hygiene, University of Rostock, Rostock, Germany
| | - Hermann Lang
- Department of Operative Dentistry and Periodontology, University of Rostock, Rostock, Germany
- * E-mail:
| |
Collapse
|
462
|
Bone marrow stromal cells as immunomodulators. A primer for dermatologists. J Dermatol Sci 2014; 77:11-20. [PMID: 25476233 DOI: 10.1016/j.jdermsci.2014.10.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 09/29/2014] [Accepted: 10/15/2014] [Indexed: 02/07/2023]
Abstract
Bone marrow stromal cells (BMSCs, also known as mesenchymal stem cells or MSCs) represent a unique cell population in the bone marrow with a long-known function to support hematopoiesis and replace skeletal tissues. The recent discovery that BMSCs also possess potent immunoregulatory features attracted a great deal of attention from stem cell biologists, immunologists and clinicians of different specialties worldwide. Initial clinical experience along with several animal models suggested that intravenously delivered BMSCs are able to regulate a wide variety of host immune cells and act in a way that is beneficial for the recipient in a variety of diseases. The role of the present review is to give a short introduction to the biology of BMSCs and to summarize our current understanding of how BMSCs modulate the immune system with special emphasis on available clinical data. Considering the audience of this journal we will also attempt to guide dermatologists in choosing the right skin conditions where BMSCs might be considered as a therapeutic alternative.
Collapse
|
463
|
Walter J, Ware LB, Matthay MA. Mesenchymal stem cells: mechanisms of potential therapeutic benefit in ARDS and sepsis. THE LANCET RESPIRATORY MEDICINE 2014; 2:1016-26. [PMID: 25465643 DOI: 10.1016/s2213-2600(14)70217-6] [Citation(s) in RCA: 187] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Multipotent mesenchymal stem (stromal) cells (MSCs) have shown promising therapeutic effects in preclinical models of both acute respiratory distress syndrome (ARDS) and sepsis. Although initial research focused on the ability of MSCs to engraft at sites of tissue injury, increasing evidence suggests that MSCs have their therapeutic effects through mechanisms unrelated to long-term incorporation into host tissue. One of the most compelling of these pathways is the ability of MSCs to interact with injured tissue through the release of soluble bioactive factors. This Review provides an overview of the general properties of MSCs, and then outlines ways in which the paracrine effects of MSCs might reduce lung injury and enhance lung repair in ARDS and sepsis. Finally, we summarise ongoing challenges in MSC research and identify areas in which the discipline might progress in the coming years.
Collapse
Affiliation(s)
- James Walter
- Departments of Medicine and Anaesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Lorraine B Ware
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Michael A Matthay
- Departments of Medicine and Anaesthesia, Cardiovascular Research Institute, University of California, San Francisco, CA, USA.
| |
Collapse
|
464
|
Abstract
PURPOSE OF REVIEW Acute respiratory distress syndrome (ARDS) is a multifaceted lung disease with no current effective therapy. Many clinical trials using conventional pharmacologic therapies have failed, suggesting the need to examine alternative approaches. Thus, attention has focused on the therapeutic potential of cell-based therapies for ARDS, with promising results demonstrated in relevant preclinical disease models. We review data concerning the therapeutic promise of cell-based therapies for ARDS. RECENT FINDINGS Recent experimental studies provide further evidence for the potential of cell-based therapies in ARDS. A number of cell types, particularly mesenchymal stem/stromal cells (MSCs), bone marrow-derived mononuclear cells, endothelial progenitor cells, and embryonic stem cells have been demonstrated to reduce mortality and modulate the inflammatory and remodeling processes in relevant preclinical ARDS models. Multiple insights have emerged in regard to the mechanisms by which cell therapies - particularly MSCs - exert their effects, with evidence supporting direct cell-mediated and paracrine-mediated mechanisms of action. Diverse paracrine mechanisms exist, including the release of cytokines, growth factors (such as keratinocyte growth factor), and antimicrobial peptides, and transfer of cellular contents such as peptides, nucleic acids, and mitochondria via either microvesicular or direct cell-cell contact-mediated transfer. SUMMARY Cell-based therapies offer considerable promise for the treatment of ARDS. While MSC-based therapies are being rapidly advanced toward clinical testing, clear therapeutic potential exists for other cell types for ARDS. A greater understanding of current knowledge gaps should further enhance the therapeutic potential of cell-based therapies for ARDS.
Collapse
|
465
|
Mao SZ, Ye X, Liu G, Song D, Liu SF. An obligatory role of NF-κB in mediating bone marrow derived endothelial progenitor cell recruitment and proliferation following endotoxemic multiple organ injury in mice. PLoS One 2014; 9:e111087. [PMID: 25333282 PMCID: PMC4205081 DOI: 10.1371/journal.pone.0111087] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 09/25/2014] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Recruitment of bone marrow derived endothelial progenitor cells (BMDEPCs) alleviates multiple organ injury (MOI) and improves outcomes. However, mechanisms mediating BMDEPC recruitment following septic MOI remain largely unknown. This study characterized the kinetics of BMDEPC recruitment and proliferation and defined the role of NF-κB in regulating BMDEPC recruitment and proliferation. METHODS AND MAIN FINDINGS Chimeric mice with an intact or disrupted NF-κB p50 gene and BMDEPC-restricted expression of green fluorescent protein were created and injected with LPS (2 mg/kg, i.p.). BMDEPC recruitment and proliferation in multiple organs were quantified. BMDEPC recruitment and proliferation are highly organ-dependent. Lungs had the highest number of BMDEPC recruitment, whereas heart, liver and kidney had only a small fraction of the number of BMDEPCs in lungs. Number of proliferating BMDEPCs was several-fold higher in lungs than in other 3 organs. Kinetically, BMDEPC recruitment into different organs showed different time course profiles. NF-κB plays obligatory roles in mediating BMDEPC recruitment and proliferation. Universal deletion of NF-κB p50 gene inhibited LPS-induced BMDEPC recruitment and proliferation by 95% and 69% in heart. However, the contribution of NF-κB to these regulations varies significantly between organs. In liver, universal p50 gene deletion reduced LPS-induced BMDEPC recruitment and proliferation only by 49% and 35%. NF-κB activities in different tissue compartments play distinct roles. Selective p50 gene deletion either in stromal/parenchymal cells or in BM/blood cells inhibited BMDEPC recruitment by a similar extent. However, selective p50 gene deletion in BM/blood cells inhibited, but in stromal/parenchymal cells augmented BMDEPC proliferation. CONCLUSIONS BMDEPC recruitment and proliferation display different kinetics in different organs following endotoxemic MOI. NF-κB plays obligatory and organ-dependent roles in regulating BMDEPC recruitment and proliferation. NF-κB activities in different tissue compartments play distinct roles in regulating BMDEPC proliferation.
Collapse
Affiliation(s)
- Sun-Zhong Mao
- Centers for Heart and Lung Research and Pulmonary and Critical Care Medicine, the Feinstein Institute for Medical Research, Manhasset, New York, United States of America
- Institute of Hypoxia Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaobing Ye
- Centers for Heart and Lung Research and Pulmonary and Critical Care Medicine, the Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - Gang Liu
- Centers for Heart and Lung Research and Pulmonary and Critical Care Medicine, the Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - Dongmei Song
- Centers for Heart and Lung Research and Pulmonary and Critical Care Medicine, the Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - Shu Fang Liu
- Centers for Heart and Lung Research and Pulmonary and Critical Care Medicine, the Feinstein Institute for Medical Research, Manhasset, New York, United States of America
- Institute of Hypoxia Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
- * E-mail:
| |
Collapse
|
466
|
Abstract
OBJECTIVE Significant progress has been made in critical care medicine during the past several decades. However, the mortality rate is still high in patients with sepsis, especially with acute kidney injury (AKI). Mesenchymal stem cells (MSCs) possess an ability to ameliorate renal injury from ischemia-reperfusion, but it is still unknown whether they have the ability to reduce sepsis-associated AKI. METHODS Male C57BL/6 mice underwent cecal ligation and puncture operation to induce sepsis and then received either normal saline or MSCs (1 × 10 cells intravenously) 3 h after surgery. RESULTS Within 24 h after cecal ligation and puncture operation, the septic mice developed kidney injury and exhibited a higher mortality. Treatment with MSCs decreased serum creatinine and blood urea nitrogen levels and improved recovery of tubular function. mRNA levels of interleukin 6 (IL-6), IL-17, tumor necrosis factor α, interferon γ, CXCL1, CXCL2, CXCL5, CCL2, and CCL3 in kidney tissue were dramatically decreased after MSC treatment. Neutrophil infiltration in kidney and blood bacterial loads were attenuated after MSC injection. Moreover, mice treated with MSCs had a higher survival rate than the saline treatment group. Injected MSCs were mainly localized in the lungs, spleen, and abdominal cavity lymph node, but not in the kidneys. CONCLUSIONS Treatment with MSCs can alleviate sepsis-associated AKI and improve survival in mice with polymicrobial sepsis. These effects may be mediated by the inhibition of IL-17 secretion and balance of the proinflammatory and anti-inflammatory states. Mesenchymal stem cells may be a potential new therapeutic agent for the prevention or reduction of sepsis-associated AKI.
Collapse
|
467
|
Amorin B, Alegretti AP, Valim V, Pezzi A, Laureano AM, da Silva MAL, Wieck A, Silla L. Mesenchymal stem cell therapy and acute graft-versus-host disease: a review. Hum Cell 2014; 27:137-50. [PMID: 24903975 PMCID: PMC4186969 DOI: 10.1007/s13577-014-0095-x] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 04/07/2014] [Indexed: 12/20/2022]
Abstract
Mesenchymal stem cells (MSCs) are being widely studied as potential cell therapy agents due to their immunomodulatory properties, which have been established by in vitro studies and in several clinical trials. Within this context, mesenchymal stem cell therapy appears to hold substantial promise, particularly in the treatment of conditions involving autoimmune and inflammatory components. Nevertheless, many research findings are still contradictory, mostly due to difficulties in characterization of the effects of MSCs in vivo. The purpose of this review is to report the mechanisms underlying mesenchymal stem cell therapy for acute graft-versus-host disease, particularly with respect to immunomodulation, migration, and homing, as well as report clinical applications described in the literature.
Collapse
Affiliation(s)
- Bruna Amorin
- Cell Therapy Center, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul Brazil
- Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Ana Paula Alegretti
- Cell Therapy Center, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul Brazil
| | - Vanessa Valim
- Cell Therapy Center, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul Brazil
| | - Annelise Pezzi
- Cell Therapy Center, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul Brazil
- Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Alvaro Macedo Laureano
- Cell Therapy Center, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul Brazil
- Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Maria Aparecida Lima da Silva
- Cell Therapy Center, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul Brazil
| | - Andréa Wieck
- Cell Therapy Center, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul Brazil
| | - Lucia Silla
- Cell Therapy Center, Experimental Research Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul Brazil
- Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Department of Hematology, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul Brazil
- Hospital de Clinicas de Porto Alegre, Ramiro Barcellos, 2350, Bairro Santa Cecília, Porto Alegre, CEP 90035-903 Brazil
| |
Collapse
|
468
|
Impact of cotransplantation of mesenchymal stem cells on lung function after unrelated allogeneic hematopoietic stem cell transplantation following non-myeloablative conditioning. Transplantation 2014; 98:348-53. [PMID: 24717223 DOI: 10.1097/tp.0000000000000068] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND In the context of hematopoietic stem cell transplantation (HSCT), mesenchymal stem cells (MSC) have been used to promote engraftment and prevent graft-versus-host disease. However, in animal models, MSC were shown to cause pulmonary alterations after systemic administration. The impact of MSC infusion on lung function has not been studied in humans. The objective of the study was to investigate the impact of MSC co-infusion on lung function and airway inflammation as well as on the incidence of pulmonary infections and cytomegalovirus (CMV) reactivation after HSCT. METHODS We have prospectively followed 30 patients who underwent unrelated HSCT with MSC co-infusion after non-myeloablative conditioning (NMA). Each patient underwent detailed lung function testing (FEV1, FVC, FEV1/FVC, RV, TLC, DLCO, and KCO) and measurement of exhaled nitric oxide before HSCT and 3, 6, and 12 months posttransplant. The incidence of pulmonary infections and CMV reactivation were also monitored. This group was compared with another group of 28 patients who underwent the same type of transplantation but without MSC co-infusion. RESULTS Lung function tests did not show important modifications over time and did not differ between the MSC and control groups. There was a higher 1-year incidence of infection, particularly of fungal infections, in patients having received a MSC co-infusion. There was no difference between groups regarding the 1-year incidence of CMV reactivation. CONCLUSIONS MSC co-infusion does not induce pulmonary deterioration 1 year after HSCT with NMA conditioning. MSC appear to be safe for the lung, but close monitoring of pulmonary infections remains essential.
Collapse
|
469
|
Host defense peptides: front-line immunomodulators. Trends Immunol 2014; 35:443-50. [DOI: 10.1016/j.it.2014.07.004] [Citation(s) in RCA: 349] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 07/10/2014] [Accepted: 07/11/2014] [Indexed: 12/30/2022]
|
470
|
Gerth DJ, King B, Rabach L, Glasgold RA, Glasgold MJ. Long-term volumetric retention of autologous fat grafting processed with closed-membrane filtration. Aesthet Surg J 2014; 34:985-94. [PMID: 25028738 DOI: 10.1177/1090820x14542649] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Some practitioners have criticized the unpredictable retention associated with autologous fat transfer. Potential causes of variations in predictability include intrinsic (patient-related) or extrinsic factors, such as harvesting, processing, and graft-delivery technique. OBJECTIVES The authors sought to determine the long-term retention of autologous fat graft processed with a closed-membrane filtration system, to compare this retention with centrifuge-processed fat, and to analyze factors that affect graft retention. METHODS This was a prospective analysis of 26 female patients (representing 52 hemi-midfaces) who underwent autologous fat transfer to the midface via the closed-membrane filtration system. The Vectra 3D camera and software were employed for all photography, which was then analyzed to compare immediate preoperative images with long-term follow-up images (obtained at least 10 months postprocedure). The authors compared the findings with data from their previous study of centrifuge-processed fat grafts (historical controls). RESULTS Mean values were as follows: age, 55 years; follow-up period, 17 months; amount of autologous fat injected, 8.88 mL; absolute volume augmentation measured at the last postoperative visit, 3.71 mL; and retention, 41.2%. Results of Welch's t test, in which the membrane-filtration data were compared with the previous centrifuge data (31.8% long-term retention), showed a significant difference (P=.03). Retention in this study was significantly higher in patients younger than 55 years (53.0% vs 31% for older patients; P=.001) and lower in patients who underwent rhytidectomy (23.8% vs 47.6% for nonrhytidectomy patients; P<.001). CONCLUSIONS Autologous fat processed by closed-membrane filtration had a significantly higher long-term retention rate than did centrifuged-processed fat injected by the same surgeons. LEVEL OF EVIDENCE 3.
Collapse
Affiliation(s)
- David J Gerth
- Dr Gerth is a volunteer Assistant Professor at the University of Miami Miller School of Medicine, DeWitt Daughtry Department of Surgery, Division of Plastic, Aesthetic and Reconstructive Surgery, Miami, FloridaDr King is a facial plastic surgeon in private practice in Northampton, MassachusettsDr Rabach is a fellow in facial plastic and reconstructive surgery at Rutgers/Robert Wood Johnson University Hospital, New Brunswick, New JerseyDrs R. A. Glasgold and M. J. Glasgold are Clinical Professors, Department of Surgery, Division of Otolaryngology and Facial Plastic Surgery at Rutgers/Robert Wood Johnson University Hospital, New Brunswick, New Jersey
| | - Bethany King
- Dr Gerth is a volunteer Assistant Professor at the University of Miami Miller School of Medicine, DeWitt Daughtry Department of Surgery, Division of Plastic, Aesthetic and Reconstructive Surgery, Miami, FloridaDr King is a facial plastic surgeon in private practice in Northampton, MassachusettsDr Rabach is a fellow in facial plastic and reconstructive surgery at Rutgers/Robert Wood Johnson University Hospital, New Brunswick, New JerseyDrs R. A. Glasgold and M. J. Glasgold are Clinical Professors, Department of Surgery, Division of Otolaryngology and Facial Plastic Surgery at Rutgers/Robert Wood Johnson University Hospital, New Brunswick, New Jersey
| | - Lesley Rabach
- Dr Gerth is a volunteer Assistant Professor at the University of Miami Miller School of Medicine, DeWitt Daughtry Department of Surgery, Division of Plastic, Aesthetic and Reconstructive Surgery, Miami, FloridaDr King is a facial plastic surgeon in private practice in Northampton, MassachusettsDr Rabach is a fellow in facial plastic and reconstructive surgery at Rutgers/Robert Wood Johnson University Hospital, New Brunswick, New JerseyDrs R. A. Glasgold and M. J. Glasgold are Clinical Professors, Department of Surgery, Division of Otolaryngology and Facial Plastic Surgery at Rutgers/Robert Wood Johnson University Hospital, New Brunswick, New Jersey
| | - Robert A Glasgold
- Dr Gerth is a volunteer Assistant Professor at the University of Miami Miller School of Medicine, DeWitt Daughtry Department of Surgery, Division of Plastic, Aesthetic and Reconstructive Surgery, Miami, FloridaDr King is a facial plastic surgeon in private practice in Northampton, MassachusettsDr Rabach is a fellow in facial plastic and reconstructive surgery at Rutgers/Robert Wood Johnson University Hospital, New Brunswick, New JerseyDrs R. A. Glasgold and M. J. Glasgold are Clinical Professors, Department of Surgery, Division of Otolaryngology and Facial Plastic Surgery at Rutgers/Robert Wood Johnson University Hospital, New Brunswick, New Jersey
| | - Mark J Glasgold
- Dr Gerth is a volunteer Assistant Professor at the University of Miami Miller School of Medicine, DeWitt Daughtry Department of Surgery, Division of Plastic, Aesthetic and Reconstructive Surgery, Miami, FloridaDr King is a facial plastic surgeon in private practice in Northampton, MassachusettsDr Rabach is a fellow in facial plastic and reconstructive surgery at Rutgers/Robert Wood Johnson University Hospital, New Brunswick, New JerseyDrs R. A. Glasgold and M. J. Glasgold are Clinical Professors, Department of Surgery, Division of Otolaryngology and Facial Plastic Surgery at Rutgers/Robert Wood Johnson University Hospital, New Brunswick, New Jersey
| |
Collapse
|
471
|
Sisto F, Bonomi A, Cavicchini L, Coccè V, Scaltrito MM, Bondiolotti G, Alessandri G, Parati E, Pessina A. Human mesenchymal stromal cells can uptake and release ciprofloxacin, acquiring in vitro anti-bacterial activity. Cytotherapy 2014; 16:181-90. [PMID: 24438899 DOI: 10.1016/j.jcyt.2013.11.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 11/19/2013] [Accepted: 11/25/2013] [Indexed: 01/05/2023]
Abstract
BACKGROUND AIMS Traditional antibiotic therapy is based on the oral or systemic injection of antibiotics that are often unable to stop a deep infection (eg, osteomyelitis). We studied whether or not bone marrow stromal cells (BM-MSCs) are able to uptake and release ciprofloxacin (CPX), a fluoroquinolone considered the drug of choice for the treatment of chronic osteomyelitis because of its favorable penetration into poorly vascularized sites of infection. METHODS Human bone marrow stromal cells (BM-MSCs) were primed with CPX (BM-MSCsCPX) according to a methodology previously standardized in our laboratory for paclitaxel (PTX). The anti-microbial activity of CPX released from BM-MSCs cells (BM-MSCsCPX-CM) or supernatant from cell lysate (BM-MSCsCPX-LYS) was evaluated by agar dilution and microdilution methods on three bacterial strains (Staphylococcus aureus, Escherichia coli and Pseudomonas aeruginosa). To investigate whether or not primed cells (BM-MSCsCPX) were able to directly act on the bacterial growth, co-colture was performed by mixing E. coli suspension to an increasing number of BM-MSCsCPX. The anti-bacterial activity was determined as number of BM-MSCsCPX that completely inhibited bacterial growth. RESULTS The results demonstrated that BM-MSCsCPX are able to uptake and then release CPX in the conditioned medium. The loaded antibiotic maintains its active form throughout the process as tested on bacteria. CONCLUSIONS Our findings suggest that CPX-loaded MSCs may represent an important device for carrying and delivering CPX (and perhaps other antibiotics) into infected deep microenvironments; they could be used for local application and by systemic infusion when their homing capacity into the bone is cleared.
Collapse
Affiliation(s)
- Francesca Sisto
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Italy
| | - Arianna Bonomi
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Italy
| | - Loredana Cavicchini
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Italy
| | - Valentina Coccè
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Italy
| | | | - Gianpietro Bondiolotti
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Italy
| | | | - Eugenio Parati
- Fondazione IRCCS, Neurological Institute Carlo Besta, Milan, Italy
| | - Augusto Pessina
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Italy.
| |
Collapse
|
472
|
Gotts JE, Abbott J, Matthay MA. Influenza causes prolonged disruption of the alveolar-capillary barrier in mice unresponsive to mesenchymal stem cell therapy. Am J Physiol Lung Cell Mol Physiol 2014; 307:L395-406. [PMID: 25038188 DOI: 10.1152/ajplung.00110.2014] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Viral pneumonia is a major cause of acute respiratory distress syndrome (ARDS). Anti-inflammatory therapies for viral-induced lung injury show promise in preclinical models. Mesenchymal stem/stromal cells (MSCs) are multipotent, self-renewing cells that secrete anti-inflammatory cytokines and epithelial and endothelial growth factors. We inoculated mice intranasally with influenza A (murine-adapted Puerto Rico/8/34) or PBS, and the mice were killed at multiple time points after infection for measures of lung injury and viral load. We report that influenza induces marked, long-lasting dysfunction of the alveolar-capillary barrier peaking at 1 wk but lasting longer than 3 wk postinfection. Weight loss, commonly employed as a criterion for euthanasia (and hence "survival"), was found to be poorly predictive of the severity of lung injury at its peak; rather, persistent weight loss 11 days postinfection identified mice with impaired injury resolution. Murine and human bone marrow-derived MSCs (obtained from the National Institutes of Health repository) were then administered intravenously during the rapid phase of injury progression. Murine MSCs (mMSCs) given two times 24 h apart failed to improve weight loss, lung water, bronchoalveolar lavage inflammation, or histology. However, mMSCs prevented influenza-induced thrombocytosis and caused a modest reduction in lung viral load at day 7. Human MSCs administered intravenously showed a similar lack of efficacy. The results demonstrate that the influenza murine model bears important similarities to the slow resolution of ARDS in patients. Despite their potent therapeutic effects in many models of acute inflammation and lung injury, MSCs do not improve influenza-mediated lung injury in mice.
Collapse
Affiliation(s)
- Jeffrey E Gotts
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California
| | - Jason Abbott
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California
| | - Michael A Matthay
- Departments of Medicine and Anesthesia, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California
| |
Collapse
|
473
|
Sutton MT, Bonfield TL. Stem cells: innovations in clinical applications. Stem Cells Int 2014; 2014:516278. [PMID: 25120571 PMCID: PMC4121181 DOI: 10.1155/2014/516278] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 12/08/2013] [Accepted: 01/13/2014] [Indexed: 01/08/2023] Open
Abstract
The use of mesenchymal stem cells (MSCs) as clinical therapeutics is a relatively new avenue of study for treatment of a variety of diseases. The therapeutic impact of the MSCs is based upon their multiplicities of function and interaction with host tissues. MSCs can be anti-inflammatory, antifibrotic, antimicrobial, and regenerative, all which may improve outcomes in scenarios of damaged tissues and inflammation. Although most studies focus on utilizing MSCs to direct clinical efficacy, it is the ability to orchestrate host response in surrounding tissue that is especially unique and versatile. This orchestration of host response can be applied to a variety of clinical scenarios not only through cell-cell interactions but also through production of bioactive secreted factors. These bioactive factors include small proteins, chemokines, cytokines, and other cellular regulators. These factors have the capacity to induce angiogenesis or blood vessel development, be chemotactic, and induce cellular recruitment. MSCs also have the capacity to differentiate with the implicated environment to regenerate tissue or accommodate host tissue in a cell specific manner. The differentiation cannot only be done in vivo but also can be optimized in vitro prior to in vivo administration, potentiating the versatility of the MSCs and opening avenues for corrective therapy and cell delivery of genes. The differentiation process depends on the environment with which the MSCs are put and results in active communication between the newly administered cells host tissue. Since these properties have been identified, there are a variety of clinical trials and studies being conducted on MSCs ability to treat human disease. This review outlines the potential use of MSCs, the types of tissue, and the innovative applications of MSCs for the treatment of diseases.
Collapse
Affiliation(s)
- Morgan T. Sutton
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106-4948, USA
- Hathaway Brown School, 19600 North Park Boulevard, Shaker Heights, OH 44122, USA
| | - Tracey L. Bonfield
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106-4948, USA
| |
Collapse
|
474
|
Masterson C, O'Toole D. The mesenchymal stromal cell magic bullet finds yet another target. Stem Cell Res Ther 2014; 5:82. [PMID: 25158102 PMCID: PMC4097829 DOI: 10.1186/scrt471] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Rojas and colleagues have presented an exciting paper demonstrating yet another relevant preclinical setting in which the mesenchymal stromal cell has a potential therapeutic application. What is particularly interesting about this study is that it addresses a disease, blood-borne systemic sepsis, which has multiple complex host responses and involves a variety of disparate organs and immune cell types. Here, the authors focus on how this injury relates more specifically to the lung, with quite dramatic improvements in several assessed injury parameters. Where does this latest demonstration of mesenchymal stromal cell efficacy leave us with regard to getting these cell therapies to the acute respiratory distress syndrome patient?
Collapse
|
475
|
Liu KD, Wilson JG, Zhuo H, Caballero L, McMillan ML, Fang X, Cosgrove K, Calfee CS, Lee JW, Kangelaris KN, Gotts JE, Rogers AJ, Levitt JE, Wiener-Kronish JP, Delucchi KL, Leavitt AD, McKenna DH, Thompson BT, Matthay MA. Design and implementation of the START (STem cells for ARDS Treatment) trial, a phase 1/2 trial of human mesenchymal stem/stromal cells for the treatment of moderate-severe acute respiratory distress syndrome. Ann Intensive Care 2014; 4:22. [PMID: 25593740 PMCID: PMC4273700 DOI: 10.1186/s13613-014-0022-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 06/04/2014] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Despite advances in supportive care, moderate-severe acute respiratory distress syndrome (ARDS) is associated with high mortality rates, and novel therapies to treat this condition are needed. Compelling pre-clinical data from mouse, rat, sheep and ex vivo perfused human lung models support the use of human mesenchymal stem (stromal) cells (MSCs) as a novel intravenous therapy for the early treatment of ARDS. METHODS This article describes the study design and challenges encountered during the implementation and phase 1 component of the START (STem cells for ARDS Treatment) trial, a phase 1/2 trial of bone marrow-derived human MSCs for moderate-severe ARDS. A trial enrolling 69 subjects is planned (9 subjects in phase 1, 60 subjects in phase 2 treated with MSCs or placebo in a 2:1 ratio). RESULTS This report describes study design features that are unique to a phase 1 trial in critically ill subjects and the specific challenges of implementation of a cell-based therapy trial in the ICU. CONCLUSIONS Experience gained during the design and implementation of the START study will be useful to investigators planning future phase 1 clinical trials based in the ICU, as well as trials of cell-based therapy for other acute illnesses. TRIAL REGISTRATION CLINICAL TRIALS REGISTRATION NCT01775774 and NCT02097641.
Collapse
Affiliation(s)
- Kathleen D Liu
- Departments of Nephrology and Anesthesia, University of California, San Francisco, CA, USA
| | - Jennifer G Wilson
- Departments of Emergency Medicine and Critical Care Medicine, University of California, San Francisco, CA, USA
| | - Hanjing Zhuo
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Lizette Caballero
- Clinical Laboratories, Bone Marrow Center, University of California, San Francisco, CA, USA
| | - Melanie L McMillan
- Clinical Laboratories, Bone Marrow Center, University of California, San Francisco, CA, USA
| | - Xiaohui Fang
- Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Katherine Cosgrove
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Carolyn S Calfee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | - Jae-Woo Lee
- Department of Anesthesia, University of California, San Francisco, CA, USA
| | - Kirsten N Kangelaris
- Division of Hospital Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | - Jeffrey E Gotts
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Francisco, CA, USA
| | - Angela J Rogers
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Joseph E Levitt
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Jeanine P Wiener-Kronish
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard University, Cambridge, MA, USA
| | - Kevin L Delucchi
- Department of Psychiatry, University of California, San Francisco, CA, USA
| | - Andrew D Leavitt
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
| | - David H McKenna
- Division of Transfusion Medicine, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - B Taylor Thompson
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Michael A Matthay
- Departments of Medicine and Anesthesia and the Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| |
Collapse
|
476
|
Stem cells, cell therapies, and bioengineering in lung biology and diseases. Comprehensive review of the recent literature 2010-2012. Ann Am Thorac Soc 2014; 10:S45-97. [PMID: 23869446 DOI: 10.1513/annalsats.201304-090aw] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
A conference, "Stem Cells and Cell Therapies in Lung Biology and Lung Diseases," was held July 25 to 28, 2011 at the University of Vermont to review the current understanding of the role of stem and progenitor cells in lung repair after injury and to review the current status of cell therapy and ex vivo bioengineering approaches for lung diseases. These are rapidly expanding areas of study that provide further insight into and challenge traditional views of mechanisms of lung repair after injury and pathogenesis of several lung diseases. The goals of the conference were to summarize the current state of the field, to discuss and debate current controversies, and to identify future research directions and opportunities for basic and translational research in cell-based therapies for lung diseases. The goal of this article, which accompanies the formal conference report, is to provide a comprehensive review of the published literature in lung regenerative medicine from the last conference report through December 2012.
Collapse
|
477
|
Stem cells and cell therapies in lung biology and diseases: conference report. Ann Am Thorac Soc 2014; 10:S25-44. [PMID: 23869447 DOI: 10.1513/annalsats.201304-089aw] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
478
|
Lalu MM, Moher D, Marshall J, Fergusson D, Mei SH, Macleod M, Griffin G, Turgeon AF, Rudnicki M, Fishman J, Avey MT, Skidmore B, Grimshaw JM, Stewart DJ, Singh K, McIntyre L. Efficacy and safety of mesenchymal stromal cells in preclinical models of acute lung injury: a systematic review protocol. Syst Rev 2014; 3:48. [PMID: 24887266 PMCID: PMC4046388 DOI: 10.1186/2046-4053-3-48] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 04/04/2014] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) in humans is caused by an unchecked proinflammatory response that results in diffuse and severe lung injury, and it is associated with a mortality rate of 35 to 45%. Mesenchymal stromal cells (MSCs; 'adult stem cells') could represent a promising new therapy for this syndrome, since preclinical evidence suggests that MSCs may ameliorate lung injury. Prior to a human clinical trial, our aim is to conduct a systematic review to compare the efficacy and safety of MSC therapy versus controls in preclinical models of acute lung injury that mimic some aspects of the human ARDS. METHODS/DESIGN We will include comparative preclinical studies (randomized and non-randomized) of acute lung injury in which MSCs were administered and outcomes compared to animals given a vehicle control. The primary outcome will be death. Secondary outcomes will include the four key features of preclinical acute lung injury as defined by the American Thoracic Society consensus conference (histologic evidence of lung injury, altered alveolar capillary barrier, lung inflammatory response, and physiological dysfunction) and pathogen clearance for acute lung injury models that are caused by infection. Electronic searches of MEDLINE, Embase, BIOSIS Previews, and Web of Science will be constructed and reviewed by the Peer Review of Electronic Search Strategies (PRESS) process. Search results will be screened independently and in duplicate. Data from eligible studies will be extracted, pooled, and analyzed using random effects models. Risk of bias will be assessed using the Cochrane risk of bias tool, and individual study reporting will be assessed according to the Animal Research: Reporting of In Vivo Experiments (ARRIVE) guidelines. DISCUSSION The results of this systematic review will comprehensively summarize the safety and efficacy of MSC therapy in preclinical models of acute lung injury. Our results will help translational scientists and clinical trialists to determine whether sufficient evidence exists to perform a human clinical trial. These results may also guide future acute lung injury preclinical and clinical research.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Lauralyn McIntyre
- Department of Medicine (Division of Critical Care), University of Ottawa, Ottawa, ON, Canada.
| | | |
Collapse
|
479
|
Khoury M, Alcayaga-Miranda F, Illanes SE, Figueroa FE. The promising potential of menstrual stem cells for antenatal diagnosis and cell therapy. Front Immunol 2014; 5:205. [PMID: 24904569 PMCID: PMC4032935 DOI: 10.3389/fimmu.2014.00205] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 04/25/2014] [Indexed: 12/21/2022] Open
Abstract
Menstrual-derived stem cells (MenSCs) are a new source of mesenchymal stem cells isolated from the menstrual fluid. Currently, there is a growing interest in their clinical potential due to fact that they are multipotent, highly proliferative, and easy to obtain in a non-invasive manner. Sampling can be repeated periodically in a simplified and reproducible manner devoid of complications that no existing cell source can match. MenSCs are also free of ethical dilemmas, and display novel properties with regard to presently known adult derived stem cells. This review details their distinctive biological properties regarding immunophenotype and function, proliferation rate, differentiation potential, and paracrine effects mediated by secreted factors. Their possible role in antenatal diagnosis is also discussed. While more insight on their immunomodulatory and diagnostic properties is needed, the impact of clinical and epidemiological factors, such as age, use of contraceptives, or hormonal status still requires further investigations to properly assess their current and future use in clinical application and diagnosis.
Collapse
Affiliation(s)
- Maroun Khoury
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de Los Andes , Santiago , Chile ; Cells for Cells , Santiago , Chile ; REGENERO, Consortium in Tissue Engineering , Santiago , Chile
| | - Francisca Alcayaga-Miranda
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de Los Andes , Santiago , Chile ; Cells for Cells , Santiago , Chile
| | - Sebastián E Illanes
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de Los Andes , Santiago , Chile
| | - Fernando E Figueroa
- Laboratory of Nano-Regenerative Medicine, Faculty of Medicine, Universidad de Los Andes , Santiago , Chile
| |
Collapse
|
480
|
Boyle AJ, McNamee JJ, McAuley DF. Biological therapies in the acute respiratory distress syndrome. Expert Opin Biol Ther 2014; 14:969-81. [PMID: 24702248 DOI: 10.1517/14712598.2014.905536] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION The acute respiratory distress syndrome (ARDS) is characterised by life-threatening respiratory failure requiring mechanical ventilation, and multiple organ failure. It has a mortality of up to 30 - 45% and causes a long-term reduction in quality of life for survivors, with only approximately 50% of survivors able to return to work 12 months after hospital discharge. AREAS COVERED In this review we discuss the complex pathophysiology of ARDS, describe the mechanistic pathways implicated in the development of ARDS and how these are currently being targeted with novel biological therapies. These include therapies targeted against inflammatory cytokines, mechanisms mediating increased alveolar permeability and disordered coagulation, as well as the potential of growth factors, gene therapy and mesenchymal stem cells. EXPERT OPINION Although understanding of the pathophysiology of ARDS has improved, to date there are no effective pharmacological interventions that target a specific mechanism, with the only potentially effective therapies to date aiming to limit ventilator-associated lung injury. However, we believe that through this improved mechanistic insight and better clinical trial design, there is cautious optimism for the future of biological therapies in ARDS, and expect current and future biological compounds to provide treatment options to clinicians managing this devastating condition.
Collapse
Affiliation(s)
- Andrew James Boyle
- Queen's University Belfast, Centre for Infection and Immunity , Belfast , UK
| | | | | |
Collapse
|
481
|
Balan A, Lucchini G, Schmidt S, Schneider A, Tramsen L, Kuçi S, Meisel R, Bader P, Lehrnbecher T. Mesenchymal stromal cells in the antimicrobial host response of hematopoietic stem cell recipients with graft-versus-host disease--friends or foes? Leukemia 2014; 28:1941-8. [PMID: 24762460 DOI: 10.1038/leu.2014.127] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Revised: 02/22/2014] [Accepted: 03/21/2014] [Indexed: 12/29/2022]
Abstract
Mesenchymal stromal cells (MSCs) are multipotent cells, which exhibit broad immunosuppressive activities. Moreover, they may be administered irrespectively of human leukocyte antigen (HLA) compatibility, without inducing life-threatening immunological reactions, as they express no HLA class II and limited HLA class I antigens under resting conditions. These characteristics have made MSC an appealing candidate for cell therapy after hematopoietic stem cell transplantation (HSCT), for example, for treatment of graft-versus-host disease (GvHD) or for graft rejection prevention/treatment in allogeneic HSCT recipients. Unfortunately, information regarding the effect of MSC infusion on the host response to infectious agents is scarce, and study results on infectious complications in patients receiving MSC are conflicting. The present review focuses on the available data from in vitro studies and animal models regarding the interaction of MSC with bacterial, viral and fungal pathogens. In a clinical part, we present the current information on infectious complications in allogeneic HSCT recipients who had received MSCs as prophylaxis or treatment of GvHD disease.
Collapse
Affiliation(s)
- A Balan
- 1] Department of Pediatric Hematology and Oncology, Children's Hospital, Johann Wolfgang Goethe University, Frankfurt, Germany [2] 'Victor Babes' University of Medicine and Pharmacy, Timisoara, Romania
| | - G Lucchini
- Department of Pediatric Hematology and Oncology, Children's Hospital, Johann Wolfgang Goethe University, Frankfurt, Germany
| | - S Schmidt
- Department of Pediatric Hematology and Oncology, Children's Hospital, Johann Wolfgang Goethe University, Frankfurt, Germany
| | - A Schneider
- Department of Pediatric Hematology and Oncology, Children's Hospital, Johann Wolfgang Goethe University, Frankfurt, Germany
| | - L Tramsen
- Department of Pediatric Hematology and Oncology, Children's Hospital, Johann Wolfgang Goethe University, Frankfurt, Germany
| | - S Kuçi
- Department of Pediatric Hematology and Oncology, Children's Hospital, Johann Wolfgang Goethe University, Frankfurt, Germany
| | - R Meisel
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Heinrich-Heine University, Düsseldorf, Germany
| | - P Bader
- Department of Pediatric Hematology and Oncology, Children's Hospital, Johann Wolfgang Goethe University, Frankfurt, Germany
| | - T Lehrnbecher
- Department of Pediatric Hematology and Oncology, Children's Hospital, Johann Wolfgang Goethe University, Frankfurt, Germany
| |
Collapse
|
482
|
Laverdet B, Micallef L, Lebreton C, Mollard J, Lataillade JJ, Coulomb B, Desmoulière A. Use of mesenchymal stem cells for cutaneous repair and skin substitute elaboration. ACTA ACUST UNITED AC 2014; 62:108-17. [DOI: 10.1016/j.patbio.2014.01.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 01/31/2014] [Indexed: 12/20/2022]
|
483
|
Rojas M, Cárdenes N, Kocyildirim E, Tedrow JR, Cáceres E, Deans R, Ting A, Bermúdez C. Human adult bone marrow-derived stem cells decrease severity of lipopolysaccharide-induced acute respiratory distress syndrome in sheep. Stem Cell Res Ther 2014; 5:42. [PMID: 24670268 PMCID: PMC4055116 DOI: 10.1186/scrt430] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 03/21/2014] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION Acute respiratory distress syndrome (ARDS) is the most common cause of respiratory failure among critically ill subjects, sepsis and severe bacterial pneumonia being its most common causes. The only interventions that have proven beneficial are protective ventilation strategies and fluid conservation approaches. New therapies are needed to address this common clinical problem. Others and we have previously shown the beneficial effect of infusion of exogenous adult stem cells in different pre-clinical models of ARDS. METHODS In the present study endotoxin was infused intravenously into 14 sheep from which 6 received different doses of adult stem cells by intrabronchial delivery to evaluate the effect of stem cell therapy. RESULTS After administration of endotoxin, there was a rapid decline in oxygenation to hypoxemic values, indicative of severe-to-moderate ARDS. None of the animals treated with saline solution recovered to normal baseline values during the 6 hours that the animals were followed. In contrast, sheep treated with a dose of 40 million adult stem cells returned their levels of oxygen in their blood to baseline two hours after the cells were infused. Similarly, improvements in carbon dioxide (CO2) clearance, pulmonary vascular pressures and inflammation were observed and confirmed by histology and by the decrease in lung edema. CONCLUSIONS We concluded that instillation of adult non-hematopoietic stem cells can diminish the impact of endotoxin and accelerate recovery of oxygenation, CO2 removal and inflammation in the ovine model, making the use of adult stem cells a real alternative for future therapies for ARDS.
Collapse
|
484
|
Cytomegalovirus infection impairs immunosuppressive and antimicrobial effector functions of human multipotent mesenchymal stromal cells. Mediators Inflamm 2014; 2014:898630. [PMID: 24782599 PMCID: PMC3981523 DOI: 10.1155/2014/898630] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 02/10/2014] [Indexed: 12/31/2022] Open
Abstract
Human mesenchymal stromal cells (MSC) possess immunosuppressive and antimicrobial effects that are partly mediated by the tryptophan-catabolizing enzyme indoleamine-2,3-dioxygenase (IDO). Therefore MSC represent a promising novel cellular immunosuppressant which has the potential to control steroid-refractory acute graft versus host disease (GvHD). In addition, MSC are capable of reducing the risk of infection in patients after haematopoietic stem cell transplantation (HST). Recent data indicate that signals from the microenvironment including those from microbes may modulate MSC effector functions. As Cytomegalovirus (CMV) represents a prominent pathogen in immunocompromised hosts, especially in patients following HST, we investigated the impact of CMV infection on MSC-mediated effects on the immune system. We demonstrate that CMV-infected MSC lose their cytokine-induced immunosuppressive capacity and are no longer able to restrict microbial growth. IDO expression is substantially impaired following CMV infection of MSC and this interaction critically depends on intact virus and the number of MSC as well as the viral load. Since overt CMV infection may undermine the clinical efficacy of MSC in the treatment of GvHD in transplant patients, we recommend that patients scheduled for MSC therapy should undergo thorough evaluation for an active CMV infection and receive CMV-directed antiviral therapy prior to the administration of MSC.
Collapse
|
485
|
Zhu YG, Hao Q, Monsel A, Feng XM, Lee JW. Adult stem cells for acute lung injury: remaining questions and concerns. Respirology 2014; 18:744-56. [PMID: 23578018 DOI: 10.1111/resp.12093] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 04/02/2013] [Indexed: 12/22/2022]
Abstract
Acute lung injury (ALI) or acute respiratory distress syndrome remains a major cause of morbidity and mortality in hospitalized patients. The pathophysiology of ALI involves complex interactions between the inciting event, such as pneumonia, sepsis or aspiration, and the host immune response resulting in lung protein permeability, impaired resolution of pulmonary oedema, an intense inflammatory response in the injured alveolus and hypoxemia. In multiple preclinical studies, adult stem cells have been shown to be therapeutic due to both the ability to mitigate injury and inflammation through paracrine mechanisms and perhaps to regenerate tissue by virtue of their multi-potency. These characteristics have stimulated intensive research efforts to explore the possibility of using stem or progenitor cells for the treatment of lung injury. A variety of stem or progenitor cells have been isolated, characterized and tested experimentally in preclinical animal models of ALI. However, questions remain concerning the optimal dose, route and the adult stem or progenitor cell to use. Here, the current mechanisms underlying the therapeutic effect of stem cells in ALI as well as the questions that will arise as clinical trials for ALI are planned are reviewed.
Collapse
Affiliation(s)
- Ying-Gang Zhu
- Department of Pulmonary Medicine, Huadong Hospital, Fudan University, Shanghai, China
| | | | | | | | | |
Collapse
|
486
|
Liu L, Yu Y, Hou Y, Chai J, Duan H, Chu W, Zhang H, Hu Q, Du J. Human umbilical cord mesenchymal stem cells transplantation promotes cutaneous wound healing of severe burned rats. PLoS One 2014; 9:e88348. [PMID: 24586314 PMCID: PMC3930522 DOI: 10.1371/journal.pone.0088348] [Citation(s) in RCA: 145] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Accepted: 01/06/2014] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Severe burns are a common and highly lethal trauma. The key step for severe burn therapy is to promote the wound healing as early as possible, and reports indicate that mesenchymal stem cell (MSC) therapy contributes to facilitate wound healing. In this study, we investigated effect of human umbilical cord MSCs (hUC-MSCs) could on wound healing in a rat model of severe burn and its potential mechanism. METHODS Adult male Wistar rats were randomly divided into sham, burn, and burn transplanted hUC-MSCs. GFP labeled hUC-MSCs or PBS was intravenous injected into respective groups. The rate of wound closure was evaluated by Image Pro Plus. GFP-labeled hUC-MSCs were tracked by in vivo bioluminescence imaging (BLI), and human-specific DNA expression in wounds was detected by PCR. Inflammatory cells, neutrophils, macrophages, capillaries and collagen types I/III in wounds were evaluated by histochemical staining. Wound blood flow was evaluated by laser Doppler blood flow meter. The levels of proinflammatory and anti-inflammatory factors, VEGF, collagen types I/III in wounds were analyzed using an ELISA. RESULTS We found that wound healing was significantly accelerated in the hUC-MSC therapy group. The hUC-MSCs migrated into wound and remarkably decreased the quantity of infiltrated inflammatory cells and levels of IL-1, IL-6, TNF-α and increased levels of IL-10 and TSG-6 in wounds. Additionally, the neovascularization and levels of VEGF in wounds in the hUC-MSC therapy group were markedly higher than those in other control groups. The ratio of collagen types I and III in the hUC-MSC therapy group were markedly higher than that in the burn group at indicated time after transplantation. CONCLUSION The study suggests that hUC-MSCs transplantation can effectively improve wound healing in severe burned rat model. Moreover, these data might provide the theoretical foundation for the further clinical application of hUC-MSC in burn areas.
Collapse
Affiliation(s)
- Lingying Liu
- Department of Burn & Plastic Surgery, the First Affiliated Hospital of PLA General Hospital, Beijing, China
| | - Yonghui Yu
- Department of Burn & Plastic Surgery, the First Affiliated Hospital of PLA General Hospital, Beijing, China
| | - Yusen Hou
- Department of Burn & Plastic Surgery, the First Affiliated Hospital of PLA General Hospital, Beijing, China
| | - Jiake Chai
- Department of Burn & Plastic Surgery, the First Affiliated Hospital of PLA General Hospital, Beijing, China
- * E-mail:
| | - Hongjie Duan
- Department of Burn & Plastic Surgery, the First Affiliated Hospital of PLA General Hospital, Beijing, China
| | - Wanli Chu
- Department of Burn & Plastic Surgery, the First Affiliated Hospital of PLA General Hospital, Beijing, China
| | - Haijun Zhang
- Department of Burn & Plastic Surgery, the First Affiliated Hospital of PLA General Hospital, Beijing, China
| | - Quan Hu
- Department of Burn & Plastic Surgery, the First Affiliated Hospital of PLA General Hospital, Beijing, China
| | - Jundong Du
- Department of Burn & Plastic Surgery, the First Affiliated Hospital of PLA General Hospital, Beijing, China
| |
Collapse
|
487
|
Huang X, Sun K, Zhao YD, Vogel SM, Song Y, Mahmud N, Zhao YY. Human CD34+ progenitor cells freshly isolated from umbilical cord blood attenuate inflammatory lung injury following LPS challenge. PLoS One 2014; 9:e88814. [PMID: 24558433 PMCID: PMC3928308 DOI: 10.1371/journal.pone.0088814] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 01/15/2014] [Indexed: 02/06/2023] Open
Abstract
Adult stem cell-based therapy is a promising novel approach for treatment of acute lung injury. Here we investigated the therapeutic potential of freshly isolated human umbilical cord blood CD34+ progenitor cells (fCB-CD34+ cells) in a mouse model of acute lung injury. At 3 h post-lipopolysaccharide (LPS) challenge, fCB-CD34+ cells were transplanted i.v. to mice while CD34− cells or PBS were administered as controls in separate cohorts of mice. We observed that fCB-CD34+ cell treatment inhibited lung vascular injury evident by decreased lung vascular permeability. In contrast, CD34− cells had no effects on lung vascular injury. Lung inflammation determined by myeloperoxidase activity, neutrophil sequestration and expression of pro-inflammatory mediators was attenuated in fCB-CD34+ cell-treated mice at 26 h post-LPS challenge compared to PBS or CD34− cell-treated controls. Importantly, lung inflammation in fCB-CD34+ cell-treated mice was returned to normal levels as seen in basal mice at 52 h post-LPS challenge whereas PBS or CD34− cell-treated control mice exhibited persistent lung inflammation. Accordingly, fCB-CD34+ cell-treated mice exhibited a marked increase of survival rate. Employing in vivo 5-bromo-2′-deoxyuridine incorporation assay, we found a drastic induction of lung endothelial proliferation in fCB-CD34+ cell-treated mice at 52 h post-LPS compared to PBS or CD34− cell-treated controls, which contributed to restoration of vascular integrity and thereby inhibition of lung inflammation. Taken together, these data have demonstrated the protective effects of fCB-CD34+ cell on acute lung injury induced by LPS challenge, suggesting fCB-CD34+ cells are an important source of stem cells for the treatment of acute lung injury.
Collapse
Affiliation(s)
- Xiaojia Huang
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
- Department of Pharmacology, School of Medical Sciences and Laboratory Medicine, Jiangsu University, Zhenjiang, China
| | - Kai Sun
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
| | - Yidan D. Zhao
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
| | - Stephen M. Vogel
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
| | - Yuanling Song
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Nadim Mahmud
- Department of Medicine, University of Illinois College of Medicine, Chicago, Illinois, United States of America
| | - You-Yang Zhao
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
- Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
488
|
McAuley DF, Curley GF, Hamid UI, Laffey JG, Abbott J, McKenna DH, Fang X, Matthay MA, Lee JW. Clinical grade allogeneic human mesenchymal stem cells restore alveolar fluid clearance in human lungs rejected for transplantation. Am J Physiol Lung Cell Mol Physiol 2014; 306:L809-15. [PMID: 24532289 DOI: 10.1152/ajplung.00358.2013] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The lack of suitable donors for all solid-organ transplant programs is exacerbated in lung transplantation by the low utilization of potential donor lungs, due primarily to donor lung injury and dysfunction, including pulmonary edema. The current studies were designed to determine if intravenous clinical-grade human mesenchymal stem (stromal) cells (hMSCs) would be effective in restoring alveolar fluid clearance (AFC) in the human ex vivo lung perfusion model, using lungs that had been deemed unsuitable for transplantation and had been subjected to prolonged ischemic time. The human lungs were perfused with 5% albumin in a balanced electrolyte solution and oxygenated with continuous positive airway pressure. Baseline AFC was measured in the control lobe and if AFC was impaired (defined as <10%/h), the lungs received either hMSC (5 × 10(6) cells) added to the perfusate or perfusion only as a control. AFC was measured in a different lung lobe at 4 h. Intravenous hMSC restored AFC in the injured lungs to a normal level. In contrast, perfusion only did not increase AFC. This positive effect on AFC was reduced by intrabronchial administration of a neutralizing antibody to keratinocyte growth factor (KGF). Thus, intravenous allogeneic hMSCs are effective in restoring the capacity of the alveolar epithelium to remove alveolar fluid at a normal rate, suggesting that this therapy may be effective in enhancing the resolution of pulmonary edema in human lungs deemed clinically unsuitable for transplantation.
Collapse
Affiliation(s)
- D F McAuley
- Health Sciences Bldg., 97, Lisburn Rd., Belfast, Northern Ireland, BT9 7BL.
| | | | | | | | | | | | | | | | | |
Collapse
|
489
|
Shalaby SM, El-Shal AS, Abd-Allah SH, Selim AO, Selim SA, Gouda ZA, Abd El Motteleb DM, Zanfaly HE, El-Assar HM, Abdelazim S. Mesenchymal stromal cell injection protects against oxidative stress in Escherichia coli-induced acute lung injury in mice. Cytotherapy 2014; 16:764-75. [PMID: 24525173 DOI: 10.1016/j.jcyt.2013.12.006] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 12/07/2013] [Accepted: 12/12/2013] [Indexed: 01/08/2023]
Abstract
BACKGROUND AIMS Stem cells may be a promising therapy for acute respiratory distress syndrome. Recent in vivo and in vitro studies suggested that the mesenchymal stromal cells (MSCs) have anti-oxidative stress properties. We hypothesized that intravenous injection of bone marrow-derived mesenchymal stem cells (MSCs) could attenuate Escherichia coli-induced acute lung injury (ALI) in mice by controlling the oxidative stress status. METHODS Eighty mice were randomly divided into four groups: group 1 (control group) received 25 μL of saline as a vehicle; group 2 contained E coli-induced ALI mice; group 3 included mice that received MSCs before induction of ALI; group 4 included mice that received MSCs after induction of ALI. Lung samples were isolated and assayed for oxidative stress variables and histopathologic analysis. Total anti-oxidant capacity was measured in broncho-alveolar lavage. RESULTS Pre- and post-injury MSC injection increased survival, reduced pulmonary edema and attenuated lung injuries in ALI mice. Histologically, MSCs exhibited a considerable degree of preservation of the pulmonary alveolar architecture. An increase of anti-oxidant enzyme activities and a decrease of myeloperoxidase activity and malondialdehyde levels in the MSC recipient groups versus the ALI group were found. Furthermore, the total anti-oxidant capacity and reduced glutathione levels were significantly increased in MSCs recipient groups versus the ALI group. Weak +ve inducible nitric oxide synthase immuno-expression in groups that received MSCs was detected. Pre-injury MSC injection showed better effects than did post-injury MSC injection. CONCLUSIONS Systemic bone marrow-derived MSC injection was effective in modulating the oxidative stress status in E coli-induced acute lung injury in mice.
Collapse
Affiliation(s)
- Sally M Shalaby
- Medical Biochemistry Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Amal S El-Shal
- Medical Biochemistry Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Somia H Abd-Allah
- Medical Biochemistry Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Assmaa O Selim
- Histology and Cell Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Sally A Selim
- Histology and Cell Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Zienab A Gouda
- Histology and Cell Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | | | - Hala E Zanfaly
- Anesthesia and Intensive Care Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Heba M El-Assar
- Anesthesia and Intensive Care Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Shymaa Abdelazim
- Microbiology and Immunology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
490
|
Zhao X, Liu D, Gong W, Zhao G, Liu L, Yang L, Hou Y. The toll-like receptor 3 ligand, poly(I:C), improves immunosuppressive function and therapeutic effect of mesenchymal stem cells on sepsis via inhibiting MiR-143. Stem Cells 2014; 32:521-33. [PMID: 24105952 DOI: 10.1002/stem.1543] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Accepted: 08/02/2013] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) are attractive candidates for clinical therapeutic applications. Recent studies indicate MSCs express active Toll-like receptors (TLRs), but their effect on MSCs and the underlying mechanisms remain unclear. In this study, we found that, after treating human umbilical cord MSCs with various TLR ligands, only TLR3 ligand, poly(I:C), could significantly increase the expression of cyclooxygenase-2 (COX-2). Furthermore, poly(I:C) could enhance MSCs' anti-inflammatory effect on macrophages. Next, we focused on the regulatory roles of microRNAs (miRNAs) in the process of poly(I:C) activating MSCs. Our experiments indicated that miR-143 expression was significantly decreased in MSCs with poly(I:C) treatment, and the expression level of miR-143 could regulate the effect of poly(I:C) on MSCs' immunosuppressive function. Subsequent results showed that the reporter genes with putative miR-143 binding sites from the transforming growth factor-β-activated kinase-1 (TAK1) and COX-2 3' untranslated regions were downregulated in the presence of miR-143. In addition, mRNA and protein expression of TAK1 and COX-2 in MSCs was also downregulated with miR-143 overexpression, suggesting that TAK1 and COX-2 are target genes of miR-143 in MSCs. Consistent with miR-143 overexpression, TAK1 interference also attenuated MSCs' immunosuppressive function enhanced by poly(I:C). Additionally, it was shown that TLR3-activated MSCs could improve survival in cecal ligation and puncture (CLP)-induced sepsis, while miR-143 overexpression reduced the effectiveness of this therapy. These results proved that poly(I:C) improved the immunosuppressive abilities of MSCs, revealed the regulatory role of miRNAs in the process, and may provide an opportunity for potential novel therapies for sepsis.
Collapse
Affiliation(s)
- Xiaoyin Zhao
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, China
| | | | | | | | | | | | | |
Collapse
|
491
|
Kox M, Pickkers P. Reply: Mesenchymal Stromal (Stem) Cell Therapy: An Emerging Immunomodulatory Strategy for the Adjunctive Treatment of Sepsis. Am J Respir Crit Care Med 2014; 189:364-5. [DOI: 10.1164/rccm.201307-1386le] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
492
|
Liles WC, Matthay MA, dos Santos CC, Weiss DJ, Stewart DJ. Mesenchymal Stromal (Stem) Cell Therapy: An Emerging Immunomodulatory Strategy for the Adjunctive Treatment of Sepsis. Am J Respir Crit Care Med 2014; 189:363-4. [DOI: 10.1164/rccm.201307-1272le] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
493
|
Skrahin A, Ahmed RK, Ferrara G, Rane L, Poiret T, Isaikina Y, Skrahina A, Zumla A, Maeurer MJ. Autologous mesenchymal stromal cell infusion as adjunct treatment in patients with multidrug and extensively drug-resistant tuberculosis: an open-label phase 1 safety trial. THE LANCET RESPIRATORY MEDICINE 2014; 2:108-22. [PMID: 24503266 DOI: 10.1016/s2213-2600(13)70234-0] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Novel treatment options are urgently needed for multidrug-resistant (MDR) and extensively drug-resistant (XDR) tuberculosis, which are associated with immune dysfunction and poor treatment outcomes. Mesenchymal stromal cells (MSCs) are immunomodulatory and adjunct autologous treatment with bone marrow-derived MSCs might improve clinical outcome by transforming chronic inflammation into productive immune responses. Our aim was to assess the safety of infusion of autologous MSCs as an adjunct treatment in patients with tuberculosis. METHODS 30 patients with microbiologically confirmed MDR or XDR tuberculosis were treated with single-dose autologous bone marrow-derived MSCs (aimed for 1×10(6) cells per kg), within 4 weeks of the start of antituberculosis-drug treatment in a specialist centre in Minsk, Belarus. Inclusion patients were those with pulmonary tuberculosis confirmed by sputum smear microscopy, culture, or both; MDR or XDR tuberculosis confirmed by drug-susceptibility testing to first-line and second-line drugs; age older than 21 years to 65 years or younger; and absence of lesion compatible with a malignant process or ongoing tuberculosis in organs other than the lungs and pleura. In addition to the inclusion criteria, patients were excluded if they were pregnant, coinfected with HIV, or infected with hepatitis B, C, or both. The primary endpoint was safety measured by MSC-infusion related events; any tuberculosis-related event within the 6 month observation period that related to a worsening of the underlying infectious disease, measured by conversion of Mycobacterium tuberculosis culture or microscopic examination; or any adverse event defined clinically or by changes in blood haematology and biochemistry variables, measured monthly for 6 months after MSC infusion per protocol. This study is registered with the German Clinical Trials Registry, number DRKS00000763. FINDINGS The most common (grade 1 or 2) adverse events were high cholesterol levels (14 of 30 patients), nausea (11 of 30 patients), and lymphopenia or diarrhoea (ten of 30 patients). There were no serious adverse events reported. We recorded two grade 3 events that were transitory-ie, increased plasma potassium ion concentrations in one patient and a transitory grade 3 γ-glutamyltransferase elevation in another patient. INTERPRETATION MSCs as an adjunct therapy are safe and can now be explored further for the treatment of patients with MDR or XDR tuberculosis in combination with standard drug regimens. Adjunct treatment with MSCs needs to be evaluated in controlled phase 2 trials to assess effects on immune responses and clinical and microbiological outcomes.
Collapse
Affiliation(s)
- Aliaksandr Skrahin
- Republican Research and Practical Centre for Pulmonology and TB, Minsk, Belarus; Department of Intensive Care, Belarussian State Medical University, Minsk, Belarus
| | - Raija K Ahmed
- Swedish Institute for Infectious Disease Control (SMI), Solna, Sweden
| | - Giovanni Ferrara
- Section of Respiratory Diseases, Department of Internal Medicine, University of Perugia, Perugia, Italy
| | - Lalit Rane
- Department of Microbiology, Tumour and Cell Biology, MTC, Karolinska Institutet, Stockholm, Sweden
| | - Thomas Poiret
- Therapeutic Immunology (TIM), Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Yanina Isaikina
- Laboratory of Cellular Biotechnology and Cytotherapy, Belarussian Research Centre for Paediatric Oncology, Haematology and Immunology, Minsk, Belarus
| | - Alena Skrahina
- Republican Research and Practical Centre for Pulmonology and TB, Minsk, Belarus
| | - Alimuddin Zumla
- Division of Infection and Immunity, Centre for Clinical Microbiology, University College London, London, UK; National Institute of Health Research-Biomedical Research Centre, University College Hospitals NHS Foundation Trust, London, UK
| | - Markus J Maeurer
- Department of Microbiology, Tumour and Cell Biology, MTC, Karolinska Institutet, Stockholm, Sweden; Therapeutic Immunology (TIM), Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Centre for Allogeneic Stem Cell Transplantation, CAST, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
494
|
Holban AM, Bleotu C, Chifiriuc MC, Bezirtzoglou E, Lazar V. Role of Pseudomonas aeruginosa quorum sensing (QS) molecules on the viability and cytokine profile of human mesenchymal stem cells. Virulence 2014; 5:303-10. [PMID: 24398422 DOI: 10.4161/viru.27571] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Pseudomonas aeruginosa infections represent one of the major threats for injured or transplanted lungs and for their healing. Considering that the mesenchymal stem cells (MSCs) are a major tool for the regenerative medicine, including therapy of lung damaging diseases, the aim of this paper was to investigate the effects of P. aeruginosa quorum sensing signaling molecules (QSSMs) on human MSCs death signaling pathways and cytokine profile. Our data revealed that N-(3-oxododecanoyl)-L-homoserine lactone (OdDHL), N-butanoyl-L-homoserine lactone (C4-HSL), 2-heptyl-3-hydroxy-4(1H)-quinolone (PQS), and its precursor, 2-heptyl-4-quinolone (HHQ), significantly impact on several core signaling mechanisms of MSCs in a specific and time-dependent manner. Even if all tested autoinducers interfered with the MSCs apoptotic genes expression, only OdDHL and HHQ significantly promoted MSCs apoptosis, by 14- and 23-fold respectively, this aspect being confirmed by the flow cytometry assay. The tested QSSMs induced a heterogeneous cytokine profile of the treated MSCs. The level of IL-1β was increased by OdDHL, IL-8 production was stimulated by all tested autoinducers, IL-6 was modulated mostly by PQS and IL-10 by HHQ. The significant influence of the purified bacterial autoinducers on the MSCs signaling pathways may suggest that the accumulation of these mediators could interfere with the normal function of these cells in the human body, and eventually, impair or abolish the success of the stem cells therapy during P. aeruginosa infections.
Collapse
Affiliation(s)
- Alina-Maria Holban
- University of Bucharest; Faculty of Biology, Department of Botanic-Microbiology; Bucharest, Romania
| | - Coralia Bleotu
- St. S. Nicolau Institute of Virology; Bucharest, Romania
| | - Mariana Carmen Chifiriuc
- University of Bucharest; Faculty of Biology, Department of Botanic-Microbiology; Bucharest, Romania
| | - Eugenia Bezirtzoglou
- Department of Food Science and Technology Laboratory of Microbiology, Biotechnology and Hygiene; Faculty of Agricultural Development; Democritus University of Thrace; Orestiada, Greece
| | - Veronica Lazar
- University of Bucharest; Faculty of Biology, Department of Botanic-Microbiology; Bucharest, Romania
| |
Collapse
|
495
|
Li J, Huang S, Wu Y, Gu C, Gao D, Feng C, Wu X, Fu X. Paracrine factors from mesenchymal stem cells: a proposed therapeutic tool for acute lung injury and acute respiratory distress syndrome. Int Wound J 2013; 11:114-21. [PMID: 24373614 DOI: 10.1111/iwj.12202] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 10/24/2013] [Accepted: 11/20/2013] [Indexed: 12/22/2022] Open
Abstract
Despite extensive researches in acute lung injury (ALI) and acute respiratory distress syndrome (ARDS), current pharmacological therapies and respiratory support are still the main methods to treat patients with ALI and ARDS and the effects remain limited. Hence, innovative therapies are needed to decrease the morbidity and mortality. Because of the proven therapeutic effects in other fields, mesenchymal stem cells (MSCs) might be considered as a promising alternative to treat ALI and ARDS. Numerous documents demonstrate that MSCs can exert multiple functions, such as engraftment, differentiation and immunoregulation, but now the key researches are concentrated on paracrine factors secreted by MSCs that can mediate endothelial and epithelial permeability, increase alveolar fluid clearance and other potential mechanisms. This review aimed to review the current researches in terms of the effects of MSCs on ALI and ARDS and to analyse these paracrine factors, as well as to predict the potential directions and challenges of the application in this field.
Collapse
Affiliation(s)
- Jiwei Li
- Department of Thoracic and Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China; Key Laboratory of Wound Repair and Regeneration of PLA, The First Affiliated Hospital, General Hospital of PLA, Trauma Center of Postgraduate Medical College, Beijing, P. R. China
| | | | | | | | | | | | | | | |
Collapse
|
496
|
Nuschke A. Activity of mesenchymal stem cells in therapies for chronic skin wound healing. Organogenesis 2013; 10:29-37. [PMID: 24322872 DOI: 10.4161/org.27405] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Chronic or non-healing skin wounds present an ongoing challenge in advanced wound care, particularly as the number of patients increases while technology aimed at stimulating wound healing in these cases remains inefficient. Mesenchymal stem cells (MSCs) have proved to be an attractive cell type for various cell therapies due to their ability to differentiate into various cell lineages, multiple donor tissue types, and relative resilience in ex-vivo expansion, as well as immunomodulatory effects during transplants. More recently, these cells have been targeted for use in strategies to improve chronic wound healing in patients with diabetic ulcers or other stasis wounds. Here, we outline several mechanisms by which MSCs can improve healing outcomes in these cases, including reducing tissue inflammation, inducing angiogenesis in the wound bed, and reducing scarring following the repair process. Approaches to extend MSC life span in implant sites are also examined.
Collapse
Affiliation(s)
- Austin Nuschke
- Department of Pathology; University of Pittsburgh; Pittsburgh, PA USA
| |
Collapse
|
497
|
Pileggi A, Xu X, Tan J, Ricordi C. Mesenchymal stromal (stem) cells to improve solid organ transplant outcome: lessons from the initial clinical trials. Curr Opin Organ Transplant 2013; 18:672-81. [PMID: 24220050 PMCID: PMC4391704 DOI: 10.1097/mot.0000000000000029] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE OF REVIEW Discuss the recent progress on the clinical use of mesenchymal stromal (stem) cells (MSC) in solid organ transplantation (SOT). RECENT FINDINGS Tissue repair and immunomodulatory properties have been recognized for MSC obtained from different human tissues. MSC-based therapy has been proposed to reduce ischemia-reperfusion injury and to promote immune tolerance. The results of recent clinical trial support the safety and promising effects of autologous and allogeneic MSC in SOT. Collectively, the use of MSC in recipients of living donor kidney transplantation was associated with improved graft function, reduced rejection, ability to omit induction and/or lower maintenance immunosuppression regimen, as well as to treat rejection episodes. SUMMARY We are living in very exciting times with the implementation of novel clinical trials aimed at establishing safety, feasibility and efficacy of cellular therapies including MSC to improve SOT outcomes. The results of the initial clinical trials support the safety of MSC-based therapy and justifying cautious optimism for the immediate future.
Collapse
Affiliation(s)
- Antonello Pileggi
- Cell Transplant Center, Diabetes Research Institute, Miami, FL 33136, USA
- The DeWitt-Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Biomedical Engineering, University of Miami, Miami, FL 33136, USA
| | - Xiumin Xu
- Cell Transplant Center, Diabetes Research Institute, Miami, FL 33136, USA
| | - Jianming Tan
- Cell and Stem Cell Institute of Xiamen University, Fuzhou, Fujian 350025, P.R. China
- Affiliated Fuzhou General Hospital of Xiamen University, Fuzhou, Fujian 350025, P.R. China
| | - Camillo Ricordi
- Cell Transplant Center, Diabetes Research Institute, Miami, FL 33136, USA
- The DeWitt-Daughtry Family Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Department of Biomedical Engineering, University of Miami, Miami, FL 33136, USA
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
498
|
Mesenchymal stem cells: environmentally responsive therapeutics for regenerative medicine. Exp Mol Med 2013; 45:e54. [PMID: 24232253 PMCID: PMC3849579 DOI: 10.1038/emm.2013.94] [Citation(s) in RCA: 849] [Impact Index Per Article: 77.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 07/22/2013] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are partially defined by their ability to differentiate into tissues including bone, cartilage and adipose in vitro, but it is their trophic, paracrine and immunomodulatory functions that may have the greatest therapeutic impact in vivo. Unlike pharmaceutical treatments that deliver a single agent at a specific dose, MSCs are site regulated and secrete bioactive factors and signals at variable concentrations in response to local microenvironmental cues. Significant progress has been made in understanding the biochemical and metabolic mechanisms and feedback associated with MSC response. The anti-inflammatory and immunomodulatory capacity of MSC may be paramount in the restoration of localized or systemic conditions for normal healing and tissue regeneration. Allogeneic MSC treatments, categorized as a drug by regulatory agencies, have been widely pursued, but new studies demonstrate the efficacy of autologous MSC therapies, even for individuals affected by a disease state. Safety and regulatory concerns surrounding allogeneic cell preparations make autologous and minimally manipulated cell therapies an attractive option for many regenerative, anti-inflammatory and autoimmune applications.
Collapse
|
499
|
Abstract
Septic shock results from the dysregulation of the innate immune response following infection. Despite major advances in fundamental and clinical research, patients diagnosed with septic shock still have a poor prognostic outcome, with a mortality rate of up to 50%. Indeed, the reasons leading to septic shock are still poorly understood. First postulated 30 years ago, the general view of septic shock as an acute and overwhelming inflammatory response still prevails today. Recently, the fact that numerous clinical trials have failed to demonstrate any positive medical outcomes has caused us to question our fundamental understanding of this condition. New and sophisticated technologies now allow us to accurately profile the various stages and contributory components of the inflammatory response defining septic shock, and many studies now report a more complex inflammatory response, particularly during the early phase of sepsis. In addition, novel experimental approaches, using more clinically relevant animal models, to standardize and stratify research outcomes are now being argued for. In the present review, we discuss the most recent findings in relation to our understanding of the underlying mechanisms involved in septic shock, and highlight the attempts made to improve animal experimental models. We also review recent studies reporting promising results with two vastly different therapeutic approaches influencing the renin-angiotensin system and applying mesenchymal stem cells for clinical intervention.
Collapse
|
500
|
Brennen WN, Denmeade SR, Isaacs JT. Mesenchymal stem cells as a vector for the inflammatory prostate microenvironment. Endocr Relat Cancer 2013; 20:R269-90. [PMID: 23975882 PMCID: PMC3994592 DOI: 10.1530/erc-13-0151] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) have an inherent tropism for sites of inflammation, which are frequently present in sites of cancer, including prostatic lesions. MSCs have been defined as CD73/CD90/CD105 triple-positive cells in the absence of hematopoietic lineage markers with the ability to differentiate into multiple mesodermal lineages, including osteoblasts, adipocytes, and chondrocytes. Our group has previously demonstrated that MSCs represent between 0.01 and 1.1% of the total cells present in human prostatectomy tissue. In addition to their multi-lineage differentiation potential, MSCs are immunoprivileged in nature and have a range of immunomodulatory effects on both the innate and adaptive arms of the immune system. MSCs have been detected in an increasing array of tissues, and evidence suggests that they are likely present in perivascular niches throughout the body. These observations suggest that MSCs represent critical mediators of the overall immune response during physiological homeostasis and likely contribute to pathophysiological conditions as well. Chronic inflammation has been suggested as an initiating event and progression factor in prostate carcinogenesis, a process in which the immunosuppressive properties of MSCs may play a role. MSCs have also been shown to influence malignant progression through a variety of other mechanisms, including effects on tumor proliferation, angiogenesis, survival, and metastasis. Additionally, human bone marrow-derived MSCs have been shown to traffic to human prostate cancer xenografts in immunocompromised murine hosts. The trafficking properties and immunoprivileged status of MSCs suggest that they can be exploited as an allogeneic cell-based vector to deliver cytotoxic or diagnostic agents for therapy.
Collapse
Affiliation(s)
- W Nathaniel Brennen
- Chemical Therapeutics Program, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland 21287, USA
| | | | | |
Collapse
|