51
|
Andreeva ER, Buravkova LB. The Role of Interplay of Mesenchymal Stromal Cells and Macrophages in Physiological and Reparative Tissue Remodeling. ACTA ACUST UNITED AC 2018. [DOI: 10.1134/s0362119718010036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
52
|
McIntyre LA, Stewart DJ, Mei SHJ, Courtman D, Watpool I, Granton J, Marshall J, dos Santos C, Walley KR, Winston BW, Schlosser K, Fergusson DA. Cellular Immunotherapy for Septic Shock. A Phase I Clinical Trial. Am J Respir Crit Care Med 2018; 197:337-347. [DOI: 10.1164/rccm.201705-1006oc] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- Lauralyn A. McIntyre
- Division of Critical Care, Department of Medicine
- Department of Epidemiology and Community Medicine, and
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Duncan J. Stewart
- Department of Cell and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Shirley H. J. Mei
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - David Courtman
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Irene Watpool
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | | | - John Marshall
- Department of Surgery and Critical Care Medicine, Keenan Research Centre of the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Claudia dos Santos
- Department of Surgery and Critical Care Medicine, Keenan Research Centre of the Li Ka Shing Knowledge Institute, St. Michael’s Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Keith R. Walley
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada; and
| | - Brent W. Winston
- Department of Critical Care Medicine, Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Kenny Schlosser
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Department of Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Dean A. Fergusson
- Department of Epidemiology and Community Medicine, and
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| |
Collapse
|
53
|
Extracellular vesicles from human-induced pluripotent stem cell-derived mesenchymal stromal cells (hiPSC-MSCs) protect against renal ischemia/reperfusion injury via delivering specificity protein (SP1) and transcriptional activating of sphingosine kinase 1 and inhibiting necroptosis. Cell Death Dis 2017; 8:3200. [PMID: 29233979 PMCID: PMC5870585 DOI: 10.1038/s41419-017-0041-4] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 10/04/2017] [Accepted: 10/06/2017] [Indexed: 02/07/2023]
Abstract
Renal ischemia-reperfusion is a main cause of acute kidney injury (AKI), which is associated with high mortality. Here we show that extracellular vesicles (EVs) secreted from hiPSC-MSCs play a critical role in protection against renal I/R injury. hiPSC-MSCs-EVs can fuse with renal cells and deliver SP1 into target cells, subsequently active SK1 expression and increase S1P formation. Chromatin immunoprecipitation (ChIP) analyses and luciferase assay were used to confirm SP1 binds directly to the SK1 promoter region and promote promoter activity. Moreover, SP1 inhibition (MIT) or SK1 inhibition (SKI-II) completely abolished the renal protective effect of hiPSC-MSCs-EVs in rat I/R injury mode. However, pre-treatment of necroptosis inhibitor Nec-1 showed no difference with the administration of hiPSC-MSCs-EVs only. We then generated an SP1 knockout hiPSC-MSC cell line by CRISPR/Cas9 system and found that SP1 knockout failed to show the protective effect of hiPSC-MSCs-EVs unless restoring the level of SP1 by Ad-SP1 in vitro and in vivo. In conclusion, this study describes an anti-necroptosis effect of hiPSC-MSCs-EVs against renal I/R injury via delivering SP1 into target renal cells and intracellular activating the expression of SK1 and the generation of S1P. These findings suggest a novel mechanism for renal protection against I/R injury, and indicate a potential therapeutic approach for a variety of renal diseases and renal transplantation.
Collapse
|
54
|
Abstract
Abstract
Sepsis is a life-threatening syndrome resulting in shock and organ dysfunction stemming from a microbial infection. Sepsis has a mortality of 40% and is implicated in half of all in-hospital deaths. The host immune response to microbial infection is critical, with early-phase sepsis characterized by a hyperinflammatory immune response, whereas the later phase of sepsis is often complicated by suppression. Sepsis has no treatment, and management remains supportive.
Stem cells constitute exciting potential therapeutic agents for sepsis. In this review, we examine the rationale for stem cells in sepsis, focusing on mesenchymal stem/stromal cells, which currently demonstrate the greatest therapeutic promise. We examine the preclinical evidence base and evaluate potential mechanisms of action of these cells that are important in the setting of sepsis. We discuss early-phase clinical trials and critically appraise translational barriers to the use of mesenchymal stem/stromal cells in patients with sepsis.
Collapse
|
55
|
Perico N, Casiraghi F, Remuzzi G. Clinical Translation of Mesenchymal Stromal Cell Therapies in Nephrology. J Am Soc Nephrol 2017; 29:362-375. [PMID: 29191959 DOI: 10.1681/asn.2017070781] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Mesenchymal stromal cells have emerged as potential candidates for cell-based therapies to modulate the immune response in organ transplantation and repair tissues after acute or chronic injury. Preclinical studies have shown convincingly in rodent models that mesenchymal stromal cells can prolong solid organ graft survival and that they can induce immune tolerance, accelerate recovery from AKI, and promote functional improvement in chronic nephropathies. Multiple complex properties of the cells, including immunomodulatory, anti-inflammatory, and proregenerative effects, seem to contribute. The promising preclinical studies have encouraged investigators to explore the safety, tolerability, and efficacy of mesenchymal stromal cell-based therapy in pilot clinical trials, including those for bone marrow and solid organ transplantation, autoimmune diseases, and tissue and organ repair. Here, we review the available data on culture-expanded mesenchymal stromal cells tested in renal transplantation, AKI, and CKD. We also briefly discuss the relevant issues that must be addressed to ensure rigorous assessment of the safety and efficacy of mesenchymal stromal cell therapies to allow the translation of this research into the practice of clinical nephrology.
Collapse
Affiliation(s)
- Norberto Perico
- Department of Renal Medicine and Molecular Medicine, IRCCS, Istituto di Ricerche Farmacologiche Mario Negri, Bergamo, Italy;
| | - Federica Casiraghi
- Department of Renal Medicine and Molecular Medicine, IRCCS, Istituto di Ricerche Farmacologiche Mario Negri, Bergamo, Italy
| | - Giuseppe Remuzzi
- Department of Renal Medicine and Molecular Medicine, IRCCS, Istituto di Ricerche Farmacologiche Mario Negri, Bergamo, Italy.,Unit of Nephrology and Dialysis, Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy; and.,L. Sacco Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| |
Collapse
|
56
|
Guillamat-Prats R, Camprubí-Rimblas M, Bringué J, Tantinyà N, Artigas A. Cell therapy for the treatment of sepsis and acute respiratory distress syndrome. ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:446. [PMID: 29264363 DOI: 10.21037/atm.2017.08.28] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Sepsis and acute respiratory distress syndrome (ARDS) are life threating diseases with high mortality and morbidity in all the critical care units around the world. After decades of research, and numerous pre-clinical and clinical trials, sepsis and ARDS remain without a specific and effective pharmacotherapy and essentially the management remains supportive. In the last years cell therapies gained potential as a therapeutic treatment for ARDS and sepsis. Based on numerous pre-clinical studies, there is a growing evidence of the potential benefits of cell based therapies for the treatment of sepsis and ARDS. Several cell types are used in the last years for the treatment of both syndromes showing high efficiency. Embryonic stem cells (ESC), multipotent stem (or stromal) cells (MSC) and epithelial progenitors cells (EpPC) have been used for both diseases. Nowadays, the major part of the pre-clinical studies are using MSC, however other relevant groups are also using induced pluripotent stem cells (iPSC) for the treatment of both syndromes and alveolar type II cells for ARDS treatment. Numerous questions need further study including: determining the best source for the progenitor cells isolation, their large scale production and cryopreservation. Also, the heterogeneity of patients with sepsis and ARDS is massive, and establish a target population or the stratification of the patients will help us to determine better the therapeutic effect of these cell therapies. In this review we are going to describe briefly the different cell types, their potential sources and characteristics and mechanism of action. Here, also we elucidate the results of several pre-clicinical and clinical studies in ARDS and in sepsis and the future directions of these studies.
Collapse
Affiliation(s)
- Raquel Guillamat-Prats
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Institut d'Investigació i Innovació Parc Taulí (I3PT), Sabadell, Catalonia, Spain
| | - Marta Camprubí-Rimblas
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Institut d'Investigació i Innovació Parc Taulí (I3PT), Sabadell, Catalonia, Spain.,Universitat Autonoma de Barcelona, Bellaterra, Catalonia, Spain
| | - Josep Bringué
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Institut d'Investigació i Innovació Parc Taulí (I3PT), Sabadell, Catalonia, Spain.,Universitat Autonoma de Barcelona, Bellaterra, Catalonia, Spain
| | - Neus Tantinyà
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Institut d'Investigació i Innovació Parc Taulí (I3PT), Sabadell, Catalonia, Spain
| | - Antonio Artigas
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), Madrid, Spain.,Institut d'Investigació i Innovació Parc Taulí (I3PT), Sabadell, Catalonia, Spain.,Universitat Autonoma de Barcelona, Bellaterra, Catalonia, Spain.,Critical Care Center, Corporació Sanitària i Universitària Parc Taulí, Sabadell, Catalonia, Spain
| |
Collapse
|
57
|
Huang X, Zhang S, Li F, Zhou Y, Wang X, Fu G, Ma X. Effects of hUCB-MSCs on recovery of neurological function and TERT expression in brain tissue of rats with cerebral ischemia-reperfusion injury. Exp Ther Med 2017; 14:5843-5846. [PMID: 29285130 PMCID: PMC5740793 DOI: 10.3892/etm.2017.5274] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 09/12/2017] [Indexed: 11/13/2022] Open
Abstract
The aim of this study was to investigate and analyze the effects of human umbilical cord blood mesenchymal stem cells (hUCB-MSCs) on the recovery of neurological function and telomerase reverse transcriptase (TERT) expression in brain tissue of rats with cerebral ischemia-reperfusion injury. A total of 100 healthy adult Wistar rats were randomly divided into two groups: The control group and the observation group according to the random number table method. After the model of cerebral ischemia-reperfusion injury was established, the rats in the observation group were treated with hUCB-MSCs (10 ml/kg), while the rats in the control group were treated with saline every day. The neurological deficit score and foot fault test were evaluated at 1, 7 and 14 days after treatment, and the rats were sacrificed at 14 days to detect the expression of TERT in brain tissue. There was no significant difference in the scores of mNSS between the two groups before the model establishment (P>0.05), but there was significant differences in two groups after the operation (P<0.05). At 1 day after the operation, the mNSS score of the two groups peaked, which was decreased in the groups with the progress of treatment. The degree of decline in the observation group was significantly greater than that in the control group (P<0.05). Similarly, there was no significant difference in the number of errors between the two groups before the model establishment (P>0.05), but there was significant difference in two groups after the operation (P<0.05). At 1 day after the operation, the number of errors also peaked, which was reduced in the groups with the progress of treatment. The degree of reduction in the observation group was significantly greater than that in the control group (P<0.05). The results of H&E staining showed it had positive reaction as nucleus or cytoplasm stained brown or yellowish brown in the observation group, while it showed neuronal shrinkage, cytoplasm and nucleus yellow dye deepening in the control group as the significant positive reaction. The gray level of the TERT protein in the brain tissue of the control group was 0.458±0.052 LOD, which was significantly lower than that in the observation group with 0.983±0.056 LOD (P<0.05). In conclusion, hUCB-MSCs can effectively improve the neurological function and the expression of TERT in brain tissue of rats with cerebral ischemia-reperfusion injury, which may be helpful to reduce the ischemia-reperfusion injury of brain tissue.
Collapse
Affiliation(s)
- Xiaohui Huang
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Shuangli Zhang
- Department of Oncology, The First Hospital of Qiqihar, Qiqihar, Heilongjiang 161005, P.R. China
| | - Fuchun Li
- Department of Orthopedics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Yuyun Zhou
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Xiaohe Wang
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Guojiao Fu
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Xueling Ma
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
58
|
Laroye C, Gibot S, Reppel L, Bensoussan D. Concise Review: Mesenchymal Stromal/Stem Cells: A New Treatment for Sepsis and Septic Shock? Stem Cells 2017; 35:2331-2339. [PMID: 28856759 DOI: 10.1002/stem.2695] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/21/2017] [Accepted: 08/21/2017] [Indexed: 12/17/2022]
Abstract
Sepsis and septic shock are the leading cause of admission and mortality in non-coronary intensive care units. Currently, however, no specific treatments are available for this syndrome. Due to the failure of conventional treatments in recent years, research is focusing on innovative therapeutic agents, including cell therapy. One particular type of cell, mesenchymal stromal/stem cells (MSCs), has raised hopes for the treatment of sepsis. Indeed, their immunomodulatory properties, antimicrobial activity and capacity of protection against organ failure confer MSCs with a major advantage to treat the immune and inflammatory dysfunctions associated with sepsis and septic shock. After a brief description of the pathophysiology of sepsis and septic shock, the latest advances in the use of MSCs to treat sepsis will be presented. Stem Cells 2017;35:2331-2339.
Collapse
Affiliation(s)
- Caroline Laroye
- Unité de Thérapie Cellulaire et banque de Tissus, CHRU de Nancy, Vandœuvre-lès-Nancy, France.,INSERM, Vandœuvre-lès-Nancy, France.,UMR 7365 CNRS, Vandœuvre-lès-Nancy, France.,Université de Lorraine, Nancy, France
| | - Sébastien Gibot
- INSERM, Vandœuvre-lès-Nancy, France.,Université de Lorraine, Nancy, France.,CHRU Nancy, Service de Réanimation Médicale, Hôpital Central, Nancy, France
| | - Loïc Reppel
- Unité de Thérapie Cellulaire et banque de Tissus, CHRU de Nancy, Vandœuvre-lès-Nancy, France.,UMR 7365 CNRS, Vandœuvre-lès-Nancy, France.,Université de Lorraine, Nancy, France
| | - Danièle Bensoussan
- Unité de Thérapie Cellulaire et banque de Tissus, CHRU de Nancy, Vandœuvre-lès-Nancy, France.,UMR 7365 CNRS, Vandœuvre-lès-Nancy, France.,Université de Lorraine, Nancy, France
| |
Collapse
|
59
|
Mesenchymal Stem Cells in Sepsis and Associated Organ Dysfunction: A Promising Future or Blind Alley? Stem Cells Int 2017; 2017:7304121. [PMID: 29098010 PMCID: PMC5618761 DOI: 10.1155/2017/7304121] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 08/06/2017] [Indexed: 12/17/2022] Open
Abstract
Sepsis, newly defined as a life-threatening organ dysfunction caused by a dysregulated host response to infection, is the most common cause of death in ICUs and one of the principal causes of death worldwide. Although substantial progress has been made in the understanding of fundamental mechanisms of sepsis, translation of these advances into clinically effective therapies has been disappointing. Given the extreme complexity of sepsis pathogenesis, the paradigm “one disease, one drug” is obviously flawed and combinations of multiple targets that involve early immunomodulation and cellular protection are needed. In this context, the immune-reprogramming properties of cell-based therapy using mesenchymal stem cells (MSC) represent an emerging therapeutic strategy in sepsis and associated organ dysfunction. This article provides an update of the current knowledge regarding MSC in preclinical models of sepsis and sepsis-induced acute kidney injury. Recommendations for further translational research in this field are discussed.
Collapse
|
60
|
Johnson V, Webb T, Norman A, Coy J, Kurihara J, Regan D, Dow S. Activated Mesenchymal Stem Cells Interact with Antibiotics and Host Innate Immune Responses to Control Chronic Bacterial Infections. Sci Rep 2017; 7:9575. [PMID: 28851894 PMCID: PMC5575141 DOI: 10.1038/s41598-017-08311-4] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 06/19/2017] [Indexed: 12/26/2022] Open
Abstract
Chronic bacterial infections associated with biofilm formation are often difficult to resolve without extended courses of antibiotic therapy. Mesenchymal stem cells (MSC) exert antibacterial activity in vitro and in acute bacterial infection models, but their activity in chronic infection with biofilm models has not been previously investigated. Therefore, we studied the effects of MSC administration in mouse and dog models of chronic infections associated with biofilms. Mice with chronic Staphylococcus aureus implant infections were treated by i.v. administration of activated or non-activated MSC, with or without antibiotic therapy. The most effective treatment protocol was identified as activated MSC co-administered with antibiotic therapy. Activated MSC were found to accumulate in the wound margins several days after i.v. administration. Macrophages in infected tissues assumed an M2 phenotype, compared to untreated infections which contained predominately M1 macrophages. Bacterial killing by MSC was found to be mediated in part by secretion of cathelicidin and was significantly increased by antibiotics. Studies in pet dogs with spontaneous chronic multi drug-resistant wound infections demonstrated clearance of bacteria and wound healing following repeated i.v. administration of activated allogeneic canine MSC. Thus, systemic therapy with activated MSC may be an effective new, non-antimicrobial approach to treatment of chronic, drug-resistant infections.
Collapse
Affiliation(s)
- Valerie Johnson
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Ft. Collins, CO, USA
| | - Tracy Webb
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Ft. Collins, CO, USA
| | - Annalis Norman
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Ft. Collins, CO, USA
| | - Jonathan Coy
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Ft. Collins, CO, USA
| | - Jade Kurihara
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Ft. Collins, CO, USA
| | - Daniel Regan
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Ft. Collins, CO, USA
| | - Steven Dow
- Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Ft. Collins, CO, USA.
| |
Collapse
|
61
|
Zhu Y, Xu L, Collins JJ, Vadivel A, Cyr-Depauw C, Zhong S, Mense L, Möbius MA, Thébaud B. Human Umbilical Cord Mesenchymal Stromal Cells Improve Survival and Bacterial Clearance in Neonatal Sepsis in Rats. Stem Cells Dev 2017; 26:1054-1064. [DOI: 10.1089/scd.2016.0329] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Yueniu Zhu
- Department of Pediatric Critical Care Medicine, Xinhua Hospital affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Pediatrics, Children's Hospital of Eastern Ontario and Children's Hospital of Ontario Research Institute (CHEORI), Ottawa, Ontario, Canada
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute (OHRI), Ottawa, Ontario, Canada
| | - Liqun Xu
- Department of Pediatrics, Children's Hospital of Eastern Ontario and Children's Hospital of Ontario Research Institute (CHEORI), Ottawa, Ontario, Canada
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute (OHRI), Ottawa, Ontario, Canada
| | - Jennifer J.P. Collins
- Department of Pediatrics, Children's Hospital of Eastern Ontario and Children's Hospital of Ontario Research Institute (CHEORI), Ottawa, Ontario, Canada
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute (OHRI), Ottawa, Ontario, Canada
- Department of Cellular and Molecular Biology, University of Ottawa, Ontario, Canada
| | - Arul Vadivel
- Department of Pediatrics, Children's Hospital of Eastern Ontario and Children's Hospital of Ontario Research Institute (CHEORI), Ottawa, Ontario, Canada
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute (OHRI), Ottawa, Ontario, Canada
| | - Chanèle Cyr-Depauw
- Department of Pediatrics, Children's Hospital of Eastern Ontario and Children's Hospital of Ontario Research Institute (CHEORI), Ottawa, Ontario, Canada
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute (OHRI), Ottawa, Ontario, Canada
| | - Shumei Zhong
- Department of Pediatrics, Children's Hospital of Eastern Ontario and Children's Hospital of Ontario Research Institute (CHEORI), Ottawa, Ontario, Canada
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute (OHRI), Ottawa, Ontario, Canada
| | - Lars Mense
- Department of Pediatrics, Children's Hospital of Eastern Ontario and Children's Hospital of Ontario Research Institute (CHEORI), Ottawa, Ontario, Canada
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute (OHRI), Ottawa, Ontario, Canada
| | - Marius A. Möbius
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute (OHRI), Ottawa, Ontario, Canada
| | - Bernard Thébaud
- Department of Pediatrics, Children's Hospital of Eastern Ontario and Children's Hospital of Ontario Research Institute (CHEORI), Ottawa, Ontario, Canada
- Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute (OHRI), Ottawa, Ontario, Canada
- Department of Cellular and Molecular Biology, University of Ottawa, Ontario, Canada
| |
Collapse
|
62
|
Prockop DJ, Oh JY, Lee RH. Data against a Common Assumption: Xenogeneic Mouse Models Can Be Used to Assay Suppression of Immunity by Human MSCs. Mol Ther 2017. [PMID: 28647464 DOI: 10.1016/j.ymthe.2017.06.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Much of what we know about immunology suggests that little is to be gained from experiments in which human cells are administered to immunocompetent mice. Multiple reports have demonstrated that this common assumption does not hold for experiments with human mesenchymal stem/stromal cells (hMSCs). The data demonstrate that hMSCs can suppress immune responses to a variety of stimuli in immunocompetent mice by a range of different mechanisms that are similar to those employed by mouse MSCs. Therefore, further experiments with hMSCs in mice will make it possible to generate preclinical data that will improve both the efficacy and safety of the clinical trials with the cells that are now in progress.
Collapse
Affiliation(s)
- Darwin J Prockop
- Institute for Regenerative Medicine, College of Medicine, Texas A&M University, 1114 TAMU, 206 Olsen Boulevard, College Station, TX 77845, USA.
| | - Joo Youn Oh
- Department of Ophthalmology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 110-744, Korea; Laboratory of Ocular Regenerative Medicine and Immunology, Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 110-744, Korea
| | - Ryang Hwa Lee
- Institute for Regenerative Medicine, College of Medicine, Texas A&M University, 1114 TAMU, 206 Olsen Boulevard, College Station, TX 77845, USA
| |
Collapse
|
63
|
Saylam G, Bayır Ö, Gültekin SS, Pınarlı FA, Han Ü, Korkmaz MH, Sancaktar ME, Tatar İ, Sargon MF, Tatar EÇ. Protective/restorative Role of the Adipose Tissue-derived Mesenchymal Stem Cells on the Radioiodine-induced Salivary Gland Damage in Rats. Radiol Oncol 2017; 51:307-316. [PMID: 28959167 PMCID: PMC5611995 DOI: 10.1515/raon-2017-0022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 04/07/2017] [Indexed: 12/29/2022] Open
Abstract
Background To analyze protective/regenerative effects of adipose tissue-derived mesenchymal stem cells (ADMSC) on 131I-Radioiodine (RAI)-induced salivary gland damage in rats. Materials and Methods Study population consisted of controls (n:6) and study groups (n:54): RAI (Group 1), ADMSC (Group 2), amifostine (Group 3), RAI+amifostine (Group 4), concomitant RAI+ADMSC (Group 5) and RAI+ADMSC after 48 h (Group 6). We used light microscopy (LM), transmission electron microscopy (TEM), and salivary gland scintigraphy (SGS), and analyzed data statistically. Results We observed the homing of ADMSC in salivary glands at 1st month on LM. RAI exposure affected necrosis, periductal fibrosis, periductal sclerosis, vascular sclerosis and the total sum score were in a statistically significant manner (P < 0.05). Intragroup comparisons with LM at 1st and 6th months revealed statistically significant improvements in Group 6 (P < 0.05) but not in Groups 4 and 5. Intergroup comparisons of the total score showed that Groups 4 and 5 in 1st month and Group 6 in 6th month had the lowest values. TEM showed vacuolization, edema, and fibrosis at 1st month, and an improvement in damage in 6th month in Groups 5 and 6. SGSs revealed significant differences for the maximum secretion ratio (Smax) (P = 0.01) and the gland-to-background ratio at a maximum count (G/BGmax) (P = 0. 01) at 1st month, for G/BGmax (P = 0.01), Smax (P = 0.01) and the time to reach the maximum count ratio over the time to reach the minimum count (Tmax/Tmin) (P = 0.03) at 6th month. 1st and 6th month scans showed differences for Smax and G/BGmax (P = 0.04), but not for Tmax/Tmin (p > 0.05). We observed a significant deterioration in gland function in group 1, whereas, mild to moderate deteriorations were seen in protective treatment groups. Conclusions Our results indicated that ADMSC might play a promising role as a protective/regenerative agent against RAI-induced salivary gland dysfunction.
Collapse
Affiliation(s)
- Güleser Saylam
- University of Health Sciences, Dışkapı Yıldırım Beyazıt Training and Research Hospital, Department of Otolaryngology, Head and Neck Surgery, Ankara, Turkey
| | - Ömer Bayır
- University of Health Sciences, Dışkapı Yıldırım Beyazıt Training and Research Hospital, Department of Otolaryngology, Head and Neck Surgery, Ankara, Turkey
| | - Salih Sinan Gültekin
- University of Health Sciences, Dışkapı Yıldırım Beyazıt Training and Research Hospital, Department of Nuclear Medicine, Ankara, Turkey
- Salih Sinan Gültekin, M.D., Department of Nuclear Medicine Dışkapı Yıldırım Beyazıt Training and Research Hospital, Ankara, Turkey. Phone: + 90 312 596 20 00
| | - Ferda Alparslan Pınarlı
- University of Health Sciences, Dışkapı Yıldırım Beyazıt Training and Research Hospital, Department of Stem Cell and Genetic Diagnostic Center, Ankara, Turkey
| | - Ünsal Han
- University of Health Sciences, Dışkapı Yıldırım Beyazıt Training and Research Hospital, Department of Pathology, Ankara, Turkey
| | - Mehmet Hakan Korkmaz
- Yıldırım Beyazıt University, Faculty of Medicine, Department of Otolaryngology, Head and Neck Surgery, Ankara, Turkey
| | - Mehmet Eser Sancaktar
- Ministry of Health, Samsun Training and Reseach Hospital, Department of Otolaryngology, Head and Neck Surgery, Samsun, Turkey
| | - İlkan Tatar
- Hacettepe University, Faculty of Medicine, Department of Anatomy, Ankara, Turkey
| | - Mustafa Fevzi Sargon
- Hacettepe University, Faculty of Medicine, Department of Anatomy, Ankara, Turkey
| | - Emel Çadallı Tatar
- University of Health Sciences, Dışkapı Yıldırım Beyazıt Training and Research Hospital, Department of Otolaryngology, Head and Neck Surgery, Ankara, Turkey
| |
Collapse
|
64
|
A novel paradigm links mitochondrial dysfunction with muscle stem cell impairment in sepsis. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2546-2553. [PMID: 28456665 DOI: 10.1016/j.bbadis.2017.04.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 04/13/2017] [Accepted: 04/18/2017] [Indexed: 02/06/2023]
Abstract
Sepsis is an acute systemic inflammatory response of the body to microbial infection and a life threatening condition associated with multiple organ failure. Survivors may display long-term disability with muscle weakness that remains poorly understood. Recent data suggest that long-term myopathy in sepsis survivors is due to failure of skeletal muscle stem cells (satellite cells) to regenerate the muscle. Satellite cells impairment in the acute phase of sepsis is linked to unusual mitochondrial dysfunctions, characterized by a dramatic reduction of the mitochondrial mass and hyperactivity of residual organelles. Survivors maintain the impairment of satellite cells, including alterations of the mitochondrial DNA (mtDNA), in the long-term. This condition can be rescued by treatment with mesenchymal stem cells (MSCs) that restore mtDNA alterations and mitochondrial function in satellite cells, and in fine their regenerative potential. Injection of MSCs in turn increases the force of isolated muscle fibers and of the whole animal, and improves the survival rate. These effects occur in the context of reduced inflammation markers that also raised during sepsis. Targeting muscle stem cells mitochondria, in a context of reduced inflammation, may represent a valuable strategy to reduce morbidity and long-term impairment of the muscle upon sepsis.
Collapse
|
65
|
Cortvrindt C, Speeckaert R, Moerman A, Delanghe JR, Speeckaert MM. The role of interleukin-17A in the pathogenesis of kidney diseases. Pathology 2017; 49:247-258. [DOI: 10.1016/j.pathol.2017.01.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Revised: 12/26/2016] [Accepted: 01/19/2017] [Indexed: 01/13/2023]
|
66
|
Zorzopulos J, Opal SM, Hernando-Insúa A, Rodriguez JM, Elías F, Fló J, López RA, Chasseing NA, Lux-Lantos VA, Coronel MF, Franco R, Montaner AD, Horn DL. Immunomodulatory oligonucleotide IMT504: Effects on mesenchymal stem cells as a first-in-class immunoprotective/immunoregenerative therapy. World J Stem Cells 2017; 9:45-67. [PMID: 28396715 PMCID: PMC5368622 DOI: 10.4252/wjsc.v9.i3.45] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 10/12/2016] [Accepted: 12/19/2016] [Indexed: 02/06/2023] Open
Abstract
The immune responses of humans and animals to insults (i.e., infections, traumas, tumoral transformation and radiation) are based on an intricate network of cells and chemical messengers. Abnormally high inflammation immediately after insult or abnormally prolonged pro-inflammatory stimuli bringing about chronic inflammation can lead to life-threatening or severely debilitating diseases. Mesenchymal stem cell (MSC) transplant has proved to be an effective therapy in preclinical studies which evaluated a vast diversity of inflammatory conditions. MSCs lead to resolution of inflammation, preparation for regeneration and actual regeneration, and then ultimate return to normal baseline or homeostasis. However, in clinical trials of transplanted MSCs, the expectations of great medical benefit have not yet been fulfilled. As a practical alternative to MSC transplant, a synthetic drug with the capacity to boost endogenous MSC expansion and/or activation may also be effective. Regarding this, IMT504, the prototype of a major class of immunomodulatory oligonucleotides, induces in vivo expansion of MSCs, resulting in a marked improvement in preclinical models of neuropathic pain, osteoporosis, diabetes and sepsis. IMT504 is easily manufactured and has an excellent preclinical safety record. In the small number of patients studied thus far, IMT504 has been well-tolerated, even at very high dosage. Further clinical investigation is necessary to demonstrate the utility of IMT504 for resolution of inflammation and regeneration in a broad array of human diseases that would likely benefit from an immunoprotective/immunoregenerative therapy.
Collapse
|
67
|
The exciting “bench to bedside” journey of cell therapies for acute kidney injury and renal transplantation. J Nephrol 2017; 30:319-336. [DOI: 10.1007/s40620-017-0384-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 02/20/2017] [Indexed: 12/15/2022]
|
68
|
Huang W, Fan W, Wang Y, Han D, Li X, Li S, Li C, Xu B, Huang Y, Fu X, Cao F. Mesenchymal stem cells in alleviating sepsis-induced mice cardiac dysfunction via inhibition of mTORC1-p70S6K signal pathway. Cell Death Discov 2017; 3:16097. [PMID: 28250969 PMCID: PMC5327616 DOI: 10.1038/cddiscovery.2016.97] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 10/14/2016] [Accepted: 10/21/2016] [Indexed: 12/30/2022] Open
Abstract
Sepsis-induced cardiac dysfunction remains a major cause of morbidity and mortality in patients suffered from severe trauma. Mesenchymal stem cells (MSCs) -based treatment has been verified as a promising approach to mitigate the sepsis-induced cardiac dysfunction, but the mechanism is still ambiguous. Thus, our study was designed to explore the potential role of MSCs in sepsis-induced cardiac dysfunction. In vivo bioluminescence imaging revealed 80% acute donor cell death of bone marrow-derived MSCs (BM-MSCs) within 3 days after transplantation. However, echocardiography demonstrated that systolic function in wild-type mice group were reduced after sepsis, while the cardiac function was relatively well persevered in cardiac-conditional deletion of Raptor (component of mTORC1 complex) mice group. Raptor KO group treated with BM-MSCs appeared better cardiac function than other groups (P<0.05). In vitro cell study revealed that co-culture of H9C2 (Raptor-Knock down) and BM-MSC could attenuate the level of proinflammatory cytokines and promote the expression of anti-inflammatory cytokine accompanied by mTORC2-Akt activation (P<0.05). In contrast, co-culture H9C2 (Raptor-O.E) and BM-MSC could aggravate the inflammatory response accompanied by the activation of mTORC1-p70S6K and inhibition of mTORC2-Akt (P<0.05). The immunomodulatory property of MSC is related to the inhibition of mTORC1-p70S6K and activation of mTORC2-Akt signaling pathway. mTORC1-p70S6K and mTORC2-Akt pathways were involved in the therapeutic adjuncts of MSC. The possible mechanism due to MSC`s immunomodulatory property through activation of mTORC2-Akt and inhibition of mTORC1-p70S6K signal pathways which may lead to modulate the expression of inflammation cytokines.
Collapse
Affiliation(s)
- Wei Huang
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China; Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Shanxi, China
| | - Wensi Fan
- Department of Cardiology, Chinese PLA General Hospital, Beijing, China; Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Shanxi, China
| | - Yabin Wang
- Department of Cardiology, Chinese PLA General Hospital , Beijing, China
| | - Dong Han
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University , Shanxi, China
| | - Xiujuan Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University , Shanxi, China
| | - Shuang Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University , Shanxi, China
| | - Congye Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University , Shanxi, China
| | - Bin Xu
- Department of Cardiology, Chinese PLA General Hospital , Beijing, China
| | - Yuesheng Huang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, The Third Military Medical University , Chongqing, China
| | - Xiaobin Fu
- Department of Cardiology, Chinese PLA General Hospital , Beijing, China
| | - Feng Cao
- Department of Cardiology, Chinese PLA General Hospital , Beijing, China
| |
Collapse
|
69
|
Nargesi AA, Lerman LO, Eirin A. Mesenchymal Stem Cell-derived Extracellular Vesicles for Renal Repair. Curr Gene Ther 2017; 17:29-42. [PMID: 28403795 PMCID: PMC5628022 DOI: 10.2174/1566523217666170412110724] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 03/26/2017] [Accepted: 04/05/2017] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Transplantation of autologous mesenchymal stem cells (MSCs) has been shown to attenuate renal injury and dysfunction in several animal models, and its efficacy is currently being tested in clinical trials for patients with renal disease. Accumulating evidence indicates that MSCs release extracellular vesicles (EVs) that deliver genes, microRNAs and proteins to recipient cells, acting as mediators of MSC paracrine actions. In this context, it is critical to characterize the MSC-derived EV cargo to elucidate their potential contribution to renal repair. In recent years, researchers have performed high-throughput sequencing and proteomic analysis to detect and identify genes, microRNAs, and proteins enriched in MSC-derived EVs. CONCLUSION The present review summarizes the current knowledge of the MSC-derived EV secretome to shed light into the mechanisms mediating MSC renal repair, and discusses preclinical and clinical studies testing the efficacy of MSC-derived EVs for treating renal disease.
Collapse
Affiliation(s)
| | - Lilach O. Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Alfonso Eirin
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
70
|
Lalu MM, Sullivan KJ, Mei SH, Moher D, Straus A, Fergusson DA, Stewart DJ, Jazi M, MacLeod M, Winston B, Marshall J, Hutton B, Walley KR, McIntyre L. Evaluating mesenchymal stem cell therapy for sepsis with preclinical meta-analyses prior to initiating a first-in-human trial. eLife 2016; 5. [PMID: 27870924 PMCID: PMC5153252 DOI: 10.7554/elife.17850] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 11/02/2016] [Indexed: 12/22/2022] Open
Abstract
Evaluation of preclinical evidence prior to initiating early-phase clinical studies has typically been performed by selecting individual studies in a non-systematic process that may introduce bias. Thus, in preparation for a first-in-human trial of mesenchymal stromal cells (MSCs) for septic shock, we applied systematic review methodology to evaluate all published preclinical evidence. We identified 20 controlled comparison experiments (980 animals from 18 publications) of in vivo sepsis models. Meta-analysis demonstrated that MSC treatment of preclinical sepsis significantly reduced mortality over a range of experimental conditions (odds ratio 0.27, 95% confidence interval 0.18–0.40, latest timepoint reported for each study). Risk of bias was unclear as few studies described elements such as randomization and no studies included an appropriately calculated sample size. Moreover, the presence of publication bias resulted in a ~30% overestimate of effect and threats to validity limit the strength of our conclusions. This novel prospective application of systematic review methodology serves as a template to evaluate preclinical evidence prior to initiating first-in-human clinical studies. DOI:http://dx.doi.org/10.7554/eLife.17850.001 Most attempts to transform exciting findings from laboratories into clinical treatments are unsuccessful. One reason for this may be the failure to consider all of the laboratory work that has been performed before deciding to test a treatment on patients for the first time. In particular, negative findings (that suggest that a potential new treatment is ineffective) may be overlooked. Stem cells may help to treat life-threatening infections, but this has not been tested in human patients. However, the effectiveness of stem cell treatments has been tested in animals that act as models of human infection. Before deciding to begin a clinical trial of stem cell therapy for life-threatening infections, Lalu et al. performed an exhaustive search to find all the studies in which stem cells were used to treat animal models of infection. Combining the results of all of these studies using particular analysis techniques revealed that stem cell therapy increased the survival of these animals overall. These positive effects were seen over a range of different experimental conditions (for example, when treating the animals with different doses of stem cells, or giving the doses at different times). Lalu et al. also identified some limitations with most of the laboratory studies that had tested stem cell therapy for infections. Many of the studies used animal models that may not be the best representations of humans with severe infection. In addition, many of the scientists did not report that they had used methods (such as randomization) that would generate the most confidence in their results. Despite these limitations, there was a lot of consistency in the reported results. Overall, the results support the decision to proceed to a clinical trial that tests the effectiveness of stem cells for treating human infections. More generally, Lalu et al.’s analysis demonstrates a way of considering all laboratory evidence before deciding to proceed to a first clinical trial in humans. This may help researchers to identify promising therapies to further develop, and also to identify potential failures before they are tested in patients. DOI:http://dx.doi.org/10.7554/eLife.17850.002
Collapse
Affiliation(s)
- Manoj M Lalu
- Department of Anesthesiology and Pain Medicine, The Ottawa Hospital, Ottawa, Canada.,Clinical Epidemiology Program, The Ottawa Hospital Research Institute, Ottawa, Canada.,Regenerative Medicine Program, The Ottawa Hospital Research Institute, Ottawa, Canada
| | - Katrina J Sullivan
- Clinical Epidemiology Program, The Ottawa Hospital Research Institute, Ottawa, Canada
| | - Shirley Hj Mei
- Regenerative Medicine Program, The Ottawa Hospital Research Institute, Ottawa, Canada
| | - David Moher
- Clinical Epidemiology Program, The Ottawa Hospital Research Institute, Ottawa, Canada.,School of Epidemiology, Public Health and Preventive Medicine, University of Ottawa, Ottawa, Canada
| | - Alexander Straus
- Clinical Epidemiology Program, The Ottawa Hospital Research Institute, Ottawa, Canada
| | - Dean A Fergusson
- Clinical Epidemiology Program, The Ottawa Hospital Research Institute, Ottawa, Canada
| | - Duncan J Stewart
- Regenerative Medicine Program, The Ottawa Hospital Research Institute, Ottawa, Canada.,Department of Cell and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | - Mazen Jazi
- Clinical Epidemiology Program, The Ottawa Hospital Research Institute, Ottawa, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Malcolm MacLeod
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - Brent Winston
- Department of Critical Care Medicine, University of Calgary, Calgary, Canada
| | - John Marshall
- Departments of Surgery and Critical Care Medicine, Keenan Research Centre of the Li KaShing Knowledge Institute, St. Michaels Hospital, The University of Toronto, Toronto, Canada
| | - Brian Hutton
- Clinical Epidemiology Program, The Ottawa Hospital Research Institute, Ottawa, Canada
| | - Keith R Walley
- Department of Medicine, Centre for Heart Lung Innovation, University of British Columbia, Vancouver, Canada
| | - Lauralyn McIntyre
- Clinical Epidemiology Program, The Ottawa Hospital Research Institute, Ottawa, Canada.,Department of Medicine, University of Ottawa, Ottawa, Canada
| |
Collapse
|
71
|
Liu W, Gao Y, Li H, Wang H, Ye M, Jiang G, Chen Y, Liu Y, Kong J, Liu W, Sun M, Hou M, Yu K. Intravenous transplantation of mesenchymal stromal cells has therapeutic effects in a sepsis mouse model through inhibition of septic natural killer cells. Int J Biochem Cell Biol 2016; 79:93-103. [PMID: 27521657 DOI: 10.1016/j.biocel.2016.08.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Revised: 07/30/2016] [Accepted: 08/09/2016] [Indexed: 12/23/2022]
Abstract
Transplantation of mesenchymal stromal cells is a promising strategy for treating sepsis. Natural killer cells are important in the development of sepsis, and their functions can be inhibited by mesenchymal stromal cells, we asked whether mesenchymal stromal cells exert their therapeutic effects through inhibiting the functions of natural killer cells in a septic mouse model generated with cecal ligation puncture method. Using co-cultures of cells, small interfering RNA, enzyme-linked immnuosorbent assays, fluorescence assays, western blotting, and pathological examination, we investigated the levels of inflammatory cytokines, proliferation of natural killer cells, inflammatory infiltration of important organs in mice, and activity of the Janus kinase/signal transducer and activator of transcription signaling pathway and found that mesenchymal stromal cells inhibited the function and proliferation of septic natural killer cells, increased interleukin-10 levels and increased the expression of components, such as Janus kinase 1, Janus kinase 2, and signal transducer and activator of transcription 3 in the Janus kinase/signal transducer and activator of transcription pathway both in vitro and in vivo. We conclude that mesenchymal stromal cells have their therapeutic effect in the septic mouse model through inhibiting the function and proliferation of septic natural killer cells. This biological process may involve interleukin-10 and suppressor of cytokine signaling 3 as well as other pathway components in the Janus kinase/signal transducer and activator of transcription pathway. Transplantation of mesenchymal stromal cells is an effective strategy to treat sepsis.
Collapse
Affiliation(s)
- Wenhua Liu
- Depatment of Intensive Care Unit (ICU), The Second Affiliated Hospital of Harbin Medical University, Harbin, Province Heilongjiang, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Province Heilongjiang, China
| | - Yang Gao
- Depatment of Intensive Care Unit (ICU), The Second Affiliated Hospital of Harbin Medical University, Harbin, Province Heilongjiang, China
| | - Haibo Li
- Depatment of Intensive Care Unit (ICU), The Second Affiliated Hospital of Harbin Medical University, Harbin, Province Heilongjiang, China
| | - Hongliang Wang
- Depatment of Intensive Care Unit (ICU), The Second Affiliated Hospital of Harbin Medical University, Harbin, Province Heilongjiang, China
| | - Ming Ye
- Depatment of Intensive Care Unit (ICU), The Second Affiliated Hospital of Harbin Medical University, Harbin, Province Heilongjiang, China
| | - Guihua Jiang
- Depatment of Intensive Care Unit (ICU), The Second Affiliated Hospital of Harbin Medical University, Harbin, Province Heilongjiang, China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Province Heilongjiang, China
| | - Yongsheng Chen
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Province Heilongjiang, China; Depatment of Urology, Harbin Medical University Cancer Hospital, Harbin, Province Heilongjiang, China
| | - Yang Liu
- Depatment of Intensive Care Unit (ICU), The Second Affiliated Hospital of Harbin Medical University, Harbin, Province Heilongjiang, China
| | - Junying Kong
- Depatment of Emergency, The Second Affiliated Hospital of Harbin Medical University, Harbin, Province Heilongjiang, China
| | - Wei Liu
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Province Heilongjiang, China
| | - Meng Sun
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Province Heilongjiang, China
| | - Meng Hou
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Province Heilongjiang, China; Depatment of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Province Heilongjiang, China
| | - Kaijiang Yu
- Department of intensive Care Unit (ICU), Harbin Medical University Cancer Hospital, Harbin, Province Heilongjiang, China.
| |
Collapse
|
72
|
Luo CJ, Luo F, Zhang L, Xu Y, Cai GY, Fu B, Feng Z, Sun XF, Chen XM. Knockout of interleukin-17A protects against sepsis-associated acute kidney injury. Ann Intensive Care 2016; 6:56. [PMID: 27334720 PMCID: PMC4917508 DOI: 10.1186/s13613-016-0157-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 06/05/2016] [Indexed: 12/14/2022] Open
Abstract
Background Sepsis-associated acute kidney injury (SA-AKI) is an independent risk factor for death in patients with sepsis, but treatment for it is limited. To improve the diagnosis and treatment of SA-AKI, we must first understand its pathogenesis. Recently, interleukin (IL)-17A has been shown to be associated with the pathogenesis of acute kidney injury and sepsis, but its role in SA-AKI remains unclear. Methods SA-AKI was induced in male C57BL/6 and IL-17A−/− mice using cecal ligation and puncture (CLP) operations for 24 h. Results At 7 days, only seven mice survived in the wild-type septic group, but nine survived in the IL-17A−/− septic group, corresponding to survival rates of 25 % and 45 %, respectively. At 24 h after CLP operations, both wild-type and IL-17A−/− septic mice developed kidney injury. The IL-17A−/− septic mice exhibited decreased serum creatinine and blood urea nitrogen levels and an improved acute tubular necrosis score. The IL-17A−/− septic mice exhibited decreased IL-6, interferon-γ, tumor necrosis factor-α, CXCL1, CXCL2, and CXCL5 expression in kidney tissue, but increased IL-10 expression. In addition, renal neutrophil infiltration was attenuated significantly in the IL-17A−/− septic group. Moreover, IL-17A−/− septic mice showed significantly decreased apoptosis of tubular epithelial cells, including decreased TUNEL-positive tubular cell number and cleaved caspase-3 level, compared with the wild-type CLP group. Their Bax/Bcl-2 expression ratio was also increased. Conclusions Our study demonstrates that IL-17A knockout could protect against SA-AKI. We show that IL-17A plays a pathogenic role in SA-AKI by increasing the levels of proinflammatory cytokines and chemokines, and by inducing neutrophil infiltration and apoptosis of tubular epithelial cells. Accordingly, IL-17A may be a novel target in SA-AKI. Electronic supplementary material The online version of this article (doi:10.1186/s13613-016-0157-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Cong-Juan Luo
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Shandong, 266003, People's Republic of China.,State Key Laboratory of Kidney Diseases, Department of Nephrology, Chinese PLA General Hospital and Medical School of Chinese PLA, No.28 Fuxing Road, Beijing, 100853, People's Republic of China
| | - Feng Luo
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Shandong, 266003, People's Republic of China.,Department of Cardiology, Liaocheng People's Hospital, Shandong, 252000, People's Republic of China
| | - Li Zhang
- State Key Laboratory of Kidney Diseases, Department of Nephrology, Chinese PLA General Hospital and Medical School of Chinese PLA, No.28 Fuxing Road, Beijing, 100853, People's Republic of China.
| | - Yan Xu
- Department of Nephrology, The Affiliated Hospital of Qingdao University, Shandong, 266003, People's Republic of China
| | - Guang-Yan Cai
- State Key Laboratory of Kidney Diseases, Department of Nephrology, Chinese PLA General Hospital and Medical School of Chinese PLA, No.28 Fuxing Road, Beijing, 100853, People's Republic of China
| | - Bo Fu
- State Key Laboratory of Kidney Diseases, Department of Nephrology, Chinese PLA General Hospital and Medical School of Chinese PLA, No.28 Fuxing Road, Beijing, 100853, People's Republic of China
| | - Zhe Feng
- State Key Laboratory of Kidney Diseases, Department of Nephrology, Chinese PLA General Hospital and Medical School of Chinese PLA, No.28 Fuxing Road, Beijing, 100853, People's Republic of China
| | - Xue-Feng Sun
- State Key Laboratory of Kidney Diseases, Department of Nephrology, Chinese PLA General Hospital and Medical School of Chinese PLA, No.28 Fuxing Road, Beijing, 100853, People's Republic of China
| | - Xiang-Mei Chen
- State Key Laboratory of Kidney Diseases, Department of Nephrology, Chinese PLA General Hospital and Medical School of Chinese PLA, No.28 Fuxing Road, Beijing, 100853, People's Republic of China.
| |
Collapse
|
73
|
Could stem cells be the future therapy for sepsis? Blood Rev 2016; 30:439-452. [PMID: 27297212 DOI: 10.1016/j.blre.2016.05.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 05/27/2016] [Accepted: 05/31/2016] [Indexed: 12/15/2022]
Abstract
The severity and threat of sepsis is well known, and despite several decades of research, the mortality continues to be high. Stem cells have great potential to be used in various clinical disorders. The innate ability of stem cells such as pluripotency, self-renewal makes them potential agents for therapeutic intervention. The pathophysiology of sepsis is a plethora of complex mechanisms which include the initial microbial infection, followed by "cytokine storm," endothelial dysfunction, coagulation cascade, and the late phase of apoptosis and immune paralysis which ultimately results in multiple organ dysfunction. Stem cells could potentially alter each step of this complex pathophysiology of sepsis. Multiple organ dysfunction associated with sepsis most often leads to death and stem cells have shown their ability to prevent the organ damage and improve the organ function. The possible mechanisms of therapeutic potential of stem cells in sepsis have been discussed in detail. The route of administration, dose level, and timing also play vital role in the overall effect of stem cells in sepsis.
Collapse
|
74
|
Jensen AR, Manning MM, Khaneki S, Drucker NA, Markel TA. Harvest tissue source does not alter the protective power of stromal cell therapy after intestinal ischemia and reperfusion injury. J Surg Res 2016; 204:361-370. [PMID: 27565072 DOI: 10.1016/j.jss.2016.05.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Revised: 04/29/2016] [Accepted: 05/03/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND Transplantation of mesenchymal stromal cells (MSCs) may be a novel treatment for intestinal ischemia. The optimal stromal cell source that could yield maximal protection after injury, however, has not been identified. We hypothesized that (1) MSCs would increase survival and mesenteric perfusion, preserve intestinal histologic architecture, and limit inflammation after intestinal ischemia and reperfusion (I/R) injury, and (2) MSCs harvested from different sources of tissue would have equivalent protective properties to the intestine after I/R inury. METHODS Adult male mice were anesthetized, and a midline laparotomy was performed. The intestines were eviscerated, the small bowel mesenteric root was identified, and baseline intestinal perfusion was determined using laser Doppler imaging. Intestinal ischemia was established by temporarily occluding the superior mesenteric artery for 60 min with a noncrushing clamp. After ischemia, the clamp was removed and the intestines were allowed to recover. Before abdominal closure, 2 × 10(6) human umbilical cord-derived MSCs, bone marrow-derived MSCs, or keratinocytes in 250 μL of phosphate-buffered saline vehicle were injected into the peritoneum. Animals were allowed to recover for 12 or 24 h (perfusion, histology, and inflammatory studies) or 7 d (survival studies). Survival data was analyzed using the log-rank test. Perfusion was expressed as a percentage of the baseline, and 12- and 24-h data was analyzed using one-way analysis of variance and the Student t-test. Nonparametric data was compared using the Mann-Whitney U-test. A P value of <0.05 was considered statistically significant. RESULTS All MSCs increased 7-d survival after I/R injury and were superior to vehicle and keratinocytes (P < 0.05). All MSCs increased mesenteric perfusion more than vehicle at 12 and 24 h after injury (P < 0.05). All MSCs provided superior perfusion compared with keratinocytes at 24 h after injury (P < 0.05). Administration of each MSC line improved intestinal histology after I/R injury (P < 0.05). Multiple proinflammatory chemokines were downregulated after the application of MSCs, suggesting a decreased inflammatory response after MSC therapy. CONCLUSIONS Transplantation of MSCs after intestinal I/R injury, irrespective of a tissue source, significantly increases survival and mesenteric perfusion and at the same time limits intestinal damage and inflammation. Further studies are needed to identify the mechanism that these cells use to promote improved outcomes after injury.
Collapse
Affiliation(s)
- Amanda R Jensen
- Department of Surgery, Section of Pediatric Surgery, Indianapolis, Indiana; Indiana University School of Medicine, Indianapolis, Indiana
| | - Morenci M Manning
- Department of Surgery, Section of Pediatric Surgery, Indianapolis, Indiana; Indiana University School of Medicine, Indianapolis, Indiana
| | - Sina Khaneki
- Department of Surgery, Section of Pediatric Surgery, Indianapolis, Indiana
| | - Natalie A Drucker
- Department of Surgery, Section of Pediatric Surgery, Indianapolis, Indiana; Indiana University School of Medicine, Indianapolis, Indiana
| | - Troy A Markel
- Department of Surgery, Section of Pediatric Surgery, Indianapolis, Indiana; Indiana University School of Medicine, Indianapolis, Indiana; Riley Hospital for Children at Indiana University Health, Indianapolis, Indiana.
| |
Collapse
|
75
|
Abstract
Kidney injury, including acute kidney injury (AKI) and chronic kidney disease (CKD), has become very common in critically ill patients treated in ICUs. Many epidemiological studies have revealed significant associations of AKI and CKD with poor outcomes of high mortality and medical costs. Although many basic studies have clarified the possible mechanisms of sepsis and septic AKI, translation of the obtained findings to clinical settings has not been successful to date. No specific drug against human sepsis or AKI is currently available. Remarkable progress of dialysis techniques such as continuous renal replacement therapy (CRRT) has enabled control of “uremia” in hemodynamically unstable patients; however, dialysis-requiring septic AKI patients are still showing unacceptably high mortality of 60–80 %. Therefore, further investigations must be conducted to improve the outcome of sepsis and septic AKI. A possible target will be remote organ injury caused by AKI. Recent basic studies have identified interleukin-6 and high mobility group box 1 (HMGB1) as important mediators for acute lung injury induced by AKI. Another target is the disease pathway that is amplified by pre-existing CKD. Vascular endothelial growth factor and HMGB1 elevations in sepsis were demonstrated to be amplified by CKD in CKD-sepsis animal models. Understanding the role of kidney injury as an amplifier in sepsis and multiple organ failure might support the identification of new drug targets for sepsis and septic AKI.
Collapse
Affiliation(s)
- Kent Doi
- Department of Emergency and Critical Care Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo, Tokyo, 113-8655 Japan
| |
Collapse
|
76
|
Bravo B, Gallego MI, Flores AI, Bornstein R, Puente-Bedia A, Hernández J, de la Torre P, García-Zaragoza E, Perez-Tavarez R, Grande J, Ballester A, Ballester S. Restrained Th17 response and myeloid cell infiltration into the central nervous system by human decidua-derived mesenchymal stem cells during experimental autoimmune encephalomyelitis. Stem Cell Res Ther 2016; 7:43. [PMID: 26987803 PMCID: PMC4797118 DOI: 10.1186/s13287-016-0304-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 02/29/2016] [Accepted: 03/02/2016] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Multiple sclerosis is a widespread inflammatory demyelinating disease. Several immunomodulatory therapies are available, including interferon-β, glatiramer acetate, natalizumab, fingolimod, and mitoxantrone. Although useful to delay disease progression, they do not provide a definitive cure and are associated with some undesirable side-effects. Accordingly, the search for new therapeutic methods constitutes an active investigation field. The use of mesenchymal stem cells (MSCs) to modify the disease course is currently the subject of intense interest. Decidua-derived MSCs (DMSCs) are a cell population obtained from human placental extraembryonic membranes able to differentiate into the three germ layers. This study explores the therapeutic potential of DMSCs. METHODS We used the experimental autoimmune encephalomyelitis (EAE) animal model to evaluate the effect of DMSCs on clinical signs of the disease and on the presence of inflammatory infiltrates in the central nervous system. We also compared the inflammatory profile of spleen T cells from DMSC-treated mice with that of EAE control animals, and the influence of DMSCs on the in vitro definition of the Th17 phenotype. Furthermore, we analyzed the effects on the presence of some critical cell types in central nervous system infiltrates. RESULTS Preventive intraperitoneal injection of DMSCs resulted in a significant delay of external signs of EAE. In addition, treatment of animals already presenting with moderate symptoms resulted in mild EAE with reduced disease scores. Besides decreased inflammatory infiltration, diminished percentages of CD4(+)IL17(+), CD11b(+)Ly6G(+) and CD11b(+)Ly6C(+) cells were found in infiltrates of treated animals. Early immune response was mitigated, with spleen cells of DMSC-treated mice displaying low proliferative response to antigen, decreased production of interleukin (IL)-17, and increased production of the anti-inflammatory cytokines IL-4 and IL-10. Moreover, lower RORγT and higher GATA-3 expression levels were detected in DMSC-treated mice. DMSCs also showed a detrimental influence on the in vitro definition of the Th17 phenotype. CONCLUSIONS DMSCs modulated the clinical course of EAE, modified the frequency and cell composition of the central nervous system infiltrates during the disease, and mediated an impairment of Th17 phenotype establishment in favor of the Th2 subtype. These results suggest that DMSCs might provide a new cell-based therapy for the control of multiple sclerosis.
Collapse
Affiliation(s)
- Beatriz Bravo
- Instituto de Salud Carlos III, Unidad Funcional de Investigación en Enfermedades Crónicas, Laboratory of Gene Regulation, Carretera de Majadahonda-Pozuelo Km 2, 28220 Madrid, Spain
| | - Marta I. Gallego
- Instituto de Salud Carlos III, Unidad Funcional de Investigación en Enfermedades Crónicas, Laboratory of Mammary Gland Pathology, Carretera de Majadahonda-Pozuelo Km 2, 28220 Madrid, Spain
| | - Ana I. Flores
- Grupo de Medicina Regenerativa, Instituto de Investigación Hospital 12 de Octubre, Avda. Córdoba s/n, 28041 Madrid, Spain
| | - Rafael Bornstein
- Hospital Central de Cruz Roja, Servicio de Hematología y Hemoterapia, Avenida de Reina Victoria 24, 28003 Madrid, Spain
| | - Alba Puente-Bedia
- Instituto de Salud Carlos III, Unidad Funcional de Investigación en Enfermedades Crónicas, Laboratory of Gene Regulation, Carretera de Majadahonda-Pozuelo Km 2, 28220 Madrid, Spain
| | - Javier Hernández
- Instituto de Salud Carlos III, Unidad Funcional de Investigación en Enfermedades Crónicas, Laboratory of Gene Regulation, Carretera de Majadahonda-Pozuelo Km 2, 28220 Madrid, Spain
| | - Paz de la Torre
- Grupo de Medicina Regenerativa, Instituto de Investigación Hospital 12 de Octubre, Avda. Córdoba s/n, 28041 Madrid, Spain
| | - Elena García-Zaragoza
- Instituto de Salud Carlos III, Unidad Funcional de Investigación en Enfermedades Crónicas, Laboratory of Mammary Gland Pathology, Carretera de Majadahonda-Pozuelo Km 2, 28220 Madrid, Spain
| | - Raquel Perez-Tavarez
- Instituto de Salud Carlos III, Unidad Funcional de Investigación en Enfermedades Crónicas, Histology Core Unit, Carretera de Majadahonda-Pozuelo Km 2, 28220 Madrid, Spain
| | - Jesús Grande
- Grupo de Medicina Regenerativa, Instituto de Investigación Hospital 12 de Octubre, Avda. Córdoba s/n, 28041 Madrid, Spain
| | - Alicia Ballester
- Instituto de Salud Carlos III, Unidad Funcional de Investigación en Enfermedades Crónicas, Laboratory of Gene Regulation, Carretera de Majadahonda-Pozuelo Km 2, 28220 Madrid, Spain
| | - Sara Ballester
- Instituto de Salud Carlos III, Unidad Funcional de Investigación en Enfermedades Crónicas, Laboratory of Gene Regulation, Carretera de Majadahonda-Pozuelo Km 2, 28220 Madrid, Spain
| |
Collapse
|
77
|
Swaminathan S, Rosner MH, Okusa MD. Emerging therapeutic targets of sepsis-associated acute kidney injury. Semin Nephrol 2015; 35:38-54. [PMID: 25795498 DOI: 10.1016/j.semnephrol.2015.01.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Sepsis-associated acute kidney injury (SA-AKI) is linked to high morbidity and mortality. To date, singular approaches to target specific pathways known to contribute to the pathogenesis of SA-AKI have failed. Because of the complexity of the pathogenesis of SA-AKI, a reassessment necessitates integrative approaches to therapeutics of SA-AKI that include general supportive therapies such as the use of vasopressors, fluids, antimicrobials, and target-specific and time-dependent therapeutics. There has been recent progress in our understanding of the pathogenesis and treatment of SA-AKI including the temporal nature of proinflammatory and anti-inflammatory processes. In this review, we discuss the clinical and experimental basis of emerging therapeutic approaches that focus on targeting early proinflammatory and late anti-inflammatory processes, as well as therapeutics that may enhance cellular survival and recovery. Finally, we include ongoing clinical trials in sepsis.
Collapse
Affiliation(s)
- Sundararaman Swaminathan
- Division of Nephrology, Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia Health System, Charlottesville, VA
| | - Mitchell H Rosner
- Division of Nephrology, Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia Health System, Charlottesville, VA
| | - Mark D Okusa
- Division of Nephrology, Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia Health System, Charlottesville, VA.
| |
Collapse
|
78
|
Mezey É, Nemeth K. Mesenchymal stem cells and infectious diseases: Smarter than drugs. Immunol Lett 2015; 168:208-14. [DOI: 10.1016/j.imlet.2015.05.020] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 05/26/2015] [Indexed: 12/11/2022]
|
79
|
Le Blanc K, Davies LC. Mesenchymal stromal cells and the innate immune response. Immunol Lett 2015; 168:140-6. [DOI: 10.1016/j.imlet.2015.05.004] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 05/05/2015] [Accepted: 05/05/2015] [Indexed: 02/06/2023]
|
80
|
Lombardo E, Poll TVD, DelaRosa O, Dalemans W. Mesenchymal stem cells as a therapeutic tool to treat sepsis. World J Stem Cells 2015; 7:368-379. [PMID: 25815121 PMCID: PMC4369493 DOI: 10.4252/wjsc.v7.i2.368] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 11/13/2014] [Accepted: 12/17/2014] [Indexed: 02/06/2023] Open
Abstract
Sepsis is a clinical syndrome caused by a deregulated host response to an infection. Sepsis is the most frequent cause of death in hospitalized patients. Although knowledge of the pathogenesis of sepsis has increased substantially during the last decades, attempts to design effective and specific therapies targeting components of the derailed host response have failed. Therefore, there is a dramatic need for new and mechanistically alternative therapies to treat this syndrome. Based on their immunomodulatory properties, adult mesenchymal stem or stromal cells (MSCs) can be a novel therapeutic tool to treat sepsis. Indeed, MSCs reduce mortality in experimental models of sepsis by modulating the deregulated inflammatory response against bacteria through the regulation of multiple inflammatory networks, the reprogramming of macrophages and neutrophils towards a more anti-inflammatory phenotype and the release of anti-microbial peptides. This report will review the current knowledge on the effects of MSC treatment in preclinical experimental small animal models of sepsis.
Collapse
|
81
|
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
|
82
|
The regulation of inflammatory mediators in acute kidney injury via exogenous mesenchymal stem cells. Mediators Inflamm 2014; 2014:261697. [PMID: 24839354 PMCID: PMC4009277 DOI: 10.1155/2014/261697] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 03/07/2014] [Accepted: 03/20/2014] [Indexed: 12/31/2022] Open
Abstract
Acute kidney injury (AKI) remains to be an independent risk factor for mortality and morbidity. Inflammation is believed to play a major role in the pathophysiology of AKI. Exogenous mesenchymal stem cells (MSCs) are now under extensive investigation as a potential therapy for AKI. Various preclinical studies indicated the beneficial effects of MSCs in alleviating renal injury and accelerating tissue repair. However the mechanisms responsible for these effects are incompletely understood. In the recent years, anti-inflammatory/immunoregulatory properties of MSCs have become one of the important issues in the treatment of AKI. This review will summarize the current literature on the regulation of inflammatory mediators via exogenous MSCs contributing to the recovery from AKI.
Collapse
|
83
|
What’s New in Shock? February 2014. Shock 2014; 41:89-90. [DOI: 10.1097/shk.0000000000000098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|