801
|
Chow L, Johnson V, Coy J, Regan D, Dow S. Mechanisms of Immune Suppression Utilized by Canine Adipose and Bone Marrow-Derived Mesenchymal Stem Cells. Stem Cells Dev 2017; 26:374-389. [PMID: 27881051 DOI: 10.1089/scd.2016.0207] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cells (MSCs) from rodents and humans have been shown to suppress T cells by distinct primary pathways, with nitric oxide (NO)-dependent pathways dominating in rodents and indoleamine 2,3-deoxygenase (IDO)-dependent pathways dominating in humans. However, the immune suppressive pathways utilized by canine MSC have not been thoroughly studied, nor have bone marrow-derived MSC (BM-MSC) and adipose-derived MSC (Ad-MSC) been directly compared for their immune modulatory potency or pathway utilization. Therefore, canine BM-MSC and Ad-MSC were generated in vitro and their potency in suppressing T cell proliferation and cytokine production was compared, and differential gene expression. Mechanisms of T cells suppression were also investigated for both MSC types. We found that BM-MSC and Ad-MSC were roughly equivalent in terms of their ability to suppress T cell activation. However, the two MSC types used both shared and distinct biochemical pathways to suppress T cell activation. Ad-MSC utilized TGF-β signaling pathways and adenosine signaling to suppress T cell activation, whereas BM-MSC used cyclooxygenase, TGF-β and adenosine signaling pathways to suppress T cell activation. These results indicate that canine MSC are distinct from human and rodent MSC terms of their immune suppressive pathways, relying primarily on cyclooxygenase and TGF-β pathways for T cell suppression, rather than on NO or IDO-mediated pathways.
Collapse
Affiliation(s)
- Lyndah Chow
- 1 Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University , Ft. Collins, Colorado.,2 Center for Immune and Regenerative Medicine, Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University , Ft. Collins, Colorado
| | - Valerie Johnson
- 1 Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University , Ft. Collins, Colorado.,2 Center for Immune and Regenerative Medicine, Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University , Ft. Collins, Colorado
| | - Jonathan Coy
- 1 Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University , Ft. Collins, Colorado.,2 Center for Immune and Regenerative Medicine, Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University , Ft. Collins, Colorado
| | - Dan Regan
- 1 Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University , Ft. Collins, Colorado.,2 Center for Immune and Regenerative Medicine, Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University , Ft. Collins, Colorado
| | - Steven Dow
- 1 Center for Immune and Regenerative Medicine, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University , Ft. Collins, Colorado.,2 Center for Immune and Regenerative Medicine, Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University , Ft. Collins, Colorado
| |
Collapse
|
802
|
Martinez B, Peplow PV. Immunomodulators and microRNAs as neurorestorative therapy for ischemic stroke. Neural Regen Res 2017; 12:865-874. [PMID: 28761412 PMCID: PMC5514854 DOI: 10.4103/1673-5374.208540] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Most of all strokes are ischemic due to occlusion of a vessel, and comprise two main types, thrombotic and embolic. Inflammation and immune response play an important role in the outcome of ischemic stroke. Pharmaceutical and cell-based therapies with immunomodulatory properties could be of benefit in treating ischemic stroke. Possible changes in microRNAs brought about by immunomodulatory treatments may be important. The pharmaceutical studies described in this review have identified several differentially regulated miRNAs associated with disregulation of mRNA targets or the upregulation of several neuroprotective genes, thereby highlighting the potential neuroprotective roles of specific miRNAs such as miR-762, -1892, -200a, -145. MiR-124, -711, -145 are the strongly associated miRNAs predicted to mediate anti-inflammatory pathways and microglia/macrophage M2-like activation phenotype. The cell-based therapy studies reviewed have mainly utilized mesenchymal stem cells or human umbilical cord blood cells and shown to improve functional and neurological outcomes in stroke animals. MiR-145 and miR-133b were implicated in nerve cell remodeling and functional recovery after stroke. Human umbilical cord blood cells decreased proinflammatory factors and promoted M2 macrophage polarization in stroke diabetic animals.
Collapse
Affiliation(s)
- Bridget Martinez
- Department of Molecular and Cellular Biology, University of California, Merced, CA, USA
| | - Philip V Peplow
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| |
Collapse
|
803
|
Dang LTT, Bui ANT, Le-Thanh Nguyen C, Truong NC, Bui ATV, Kim NP, Truong KD, Van Pham P. Intravenous Infusion of Human Adipose Tissue-Derived Mesenchymal Stem Cells to Treat Type 1 Diabetic Mellitus in Mice: An Evaluation of Grafted Cell Doses. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1083:145-156. [PMID: 29423674 DOI: 10.1007/5584_2017_127] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Mesenchymal stem cell (MSC) transplantation is a novel treatment for diabetes mellitus, especially type 1 diabetes. Many recent publications have demonstrated the efficacy of MSC transplantation on reducing blood glucose and increasing insulin production in both preclinical and clinical trials. However, the investigation of grafted cell doses has been lacking. Therefore, this study aimed to evaluate the different doses of MSCs on treatment of type 1 diabetes in mouse models. MSCs were isolated and expanded from human adipose tissue. Streptozotocin (STZ)-induced diabetic mice were divided into two groups that were intravenously transfused with two different doses of human MSCs: 106 or 2.106 cells/mouse. After transplantation, both grafted and placebo mice were monitored weekly for their blood glucose levels, glucose and insulin tolerance, pancreatic structural changes, and insulin production for 56 days after transplantation. The results showed that the higher dose of MSCs (2.106 cells/mouse) remarkably reduced death rate. The death rates were 50%, 66%, and 0% in placebo group, low-dose (1.106 MSCs) group, and high-dose (2.106 MSCs) group, respectively, after 56 days of treatment. Moreover, blood glucose levels were lower for the high-dose group compared to other groups. Glucose and insulin tolerance, as well as insulin production, were significantly improved in mice transplanted with 2.106 cells. The histochemical analyses also support these results. Thus, a higher (e.g., 2.106) dose of MSCs may be an effective dose for treatment of type 1 diabetes mellitus.
Collapse
Affiliation(s)
- Loan Thi-Tung Dang
- Stem Cell Institute, University of Science, VNUHCM, Ho Chi Minh city, Vietnam
| | - Anh Nguyen-Tu Bui
- Stem Cell Institute, University of Science, VNUHCM, Ho Chi Minh city, Vietnam
| | - Cong Le-Thanh Nguyen
- Stem Cell Institute, University of Science, VNUHCM, Ho Chi Minh city, Vietnam
- Laboratory of Stem Cell Research and Application, University of Science, VNUHCM, Ho Chi Minh city, Vietnam
| | - Nhat Chau Truong
- Stem Cell Institute, University of Science, VNUHCM, Ho Chi Minh city, Vietnam
- Laboratory of Stem Cell Research and Application, University of Science, VNUHCM, Ho Chi Minh city, Vietnam
| | - Anh Thi-Van Bui
- Stem Cell Institute, University of Science, VNUHCM, Ho Chi Minh city, Vietnam
- Laboratory of Stem Cell Research and Application, University of Science, VNUHCM, Ho Chi Minh city, Vietnam
| | - Ngoc Phan Kim
- Stem Cell Institute, University of Science, VNUHCM, Ho Chi Minh city, Vietnam
| | | | - Phuc Van Pham
- Stem Cell Institute, University of Science, VNUHCM, Ho Chi Minh city, Vietnam.
- Laboratory of Stem Cell Research and Application, University of Science, VNUHCM, Ho Chi Minh city, Vietnam.
| |
Collapse
|
804
|
Grégoire C, Lechanteur C, Briquet A, Baudoux É, Baron F, Louis E, Beguin Y. Review article: mesenchymal stromal cell therapy for inflammatory bowel diseases. Aliment Pharmacol Ther 2017; 45:205-221. [PMID: 27878827 DOI: 10.1111/apt.13864] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 06/21/2016] [Accepted: 10/25/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND Inflammatory bowel diseases (IBD) are chronic relapsing diseases in which pro-inflammatory immune cells and cytokines induce intestinal tissue damage and disability. Mesenchymal stromal cells (MSCs) exert powerful immunomodulatory effects and stimulate tissue repair. AIM To review the current data on mesenchymal stromal cell therapy in IBD. METHOD We searched PubMed and 'ClinicalTrials.gov' databases using the terms 'mesenchymal stromal cells', 'mesenchymal stem cell transplantation', 'inflammatory bowel diseases', 'Crohn disease' and 'colitis, ulcerative'. Additional publications were identified from individual article reference lists. RESULTS MSCs include inhibition of Th1/Th17 lymphocytes and recruitment of regulatory T lymphocytes, induction of antigen-presenting cells into a regulatory-like profile, and stimulation of epithelial cell differentiation and proliferation. More than 200 patients with refractory fistulas have been treated with local injections of MSCs, resulting in complete response in more than half, and in overall response in approximately two thirds of patients. In refractory luminal Crohn's disease, 49 cases of systemic MSC infusions have been reported, while trials with autologous MSCs resulted in mitigated responses, studies using allogeneic MSCs were promising, with around 60% of patients experiencing a response and around 40% achieving clinical remission. CONCLUSIONS Mesenchymal stromal cells might represent a promising therapy for IBD, especially for Crohn's disease. There remain many unsolved questions concerning the optimal origin and source of mesenchymal stromal cells, dosage and modalities of administration. Moreover, mesenchymal stromal cells still need to prove their effectiveness compared with conventional treatments in randomised controlled trials.
Collapse
Affiliation(s)
- C Grégoire
- Unit of Haematology, Department of Haematology, CHU of Liège, GIGA-I3, University of Liège, Liège, Belgium
| | - C Lechanteur
- Laboratory of Cell and Gene Therapy (LTCG), CHU of Liège, Liège, Belgium
| | - A Briquet
- Laboratory of Cell and Gene Therapy (LTCG), CHU of Liège, Liège, Belgium
| | - É Baudoux
- Laboratory of Cell and Gene Therapy (LTCG), CHU of Liège, Liège, Belgium
| | - F Baron
- Unit of Haematology, Department of Haematology, CHU of Liège, GIGA-I3, University of Liège, Liège, Belgium
| | - E Louis
- Department of Gastroenterology, CHU of Liège, University of Liège, Liège, Belgium
| | - Y Beguin
- Unit of Haematology, Department of Haematology, CHU of Liège, GIGA-I3, University of Liège, Liège, Belgium.,Laboratory of Cell and Gene Therapy (LTCG), CHU of Liège, Liège, Belgium
| |
Collapse
|
805
|
Shirjang S, Mansoori B, Solali S, Hagh MF, Shamsasenjan K. Toll-like receptors as a key regulator of mesenchymal stem cell function: An up-to-date review. Cell Immunol 2016; 315:1-10. [PMID: 28284487 DOI: 10.1016/j.cellimm.2016.12.005] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Revised: 12/07/2016] [Accepted: 12/24/2016] [Indexed: 02/07/2023]
Abstract
Understanding the role of toll-like receptors (TLRs) in the immunomodulation potential, differentiation, migration, and survival of mesenchymal stem cells (MSCs) is absolutely vital to fully exploiting their MSC-based therapeutic potential. Furthermore, through recognition of exogenous or endogenous ligands produced upon injury, TLRs have been linked to allograft rejection and maintenance of chronic inflammatory diseases, including Crohn's disease, rheumatoid arthritis. Characterizing the effect of TLRs in biological control of MSCs fate and function could improve our knowledge about the MSC-based cell therapy and immunotherapy. In this paper, we outline the impacts of TLR activation and mechanisms on MSCs immunomodulatory functions, differentiation, migration, and survivability. Moreover, we indicate that the expression patterns of TLRs in MSCs from different sources.
Collapse
Affiliation(s)
- Solmaz Shirjang
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Solali
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Karim Shamsasenjan
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
806
|
New Role of Adult Lung c-kit + Cells in a Mouse Model of Airway Hyperresponsiveness. Mediators Inflamm 2016; 2016:3917471. [PMID: 28090152 PMCID: PMC5206449 DOI: 10.1155/2016/3917471] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 10/05/2016] [Accepted: 11/10/2016] [Indexed: 12/16/2022] Open
Abstract
Structural changes contribute to airway hyperresponsiveness and airflow obstruction in asthma. Emerging evidence points to the involvement of c-kit+ cells in lung homeostasis, although their potential role in asthma is unknown. Our aim was to isolate c-kit+ cells from normal mouse lungs and to test whether these cells can interfere with hallmarks of asthma in an animal model. Adult mouse GFP-tagged c-kit+ cells, intratracheally delivered in the ovalbumin-induced airway hyperresponsiveness, positively affected airway remodeling and improved airway function. In bronchoalveolar lavage fluid of cell-treated animals, a reduction in the number of inflammatory cells and in IL-4, IL-5, and IL-13 release, along with an increase of IL-10, was observed. In MSC-treated mice, the macrophage polarization to M2-like subset may explain, at least in part, the increment in the level of anti-inflammatory cytokine IL-10. After in vitro stimulation of c-kit+ cells with proinflammatory cytokines, the indoleamine 2,3-dioxygenase and TGFβ were upregulated. These data, together with the increased apoptosis of inflammatory cells in vivo, indicate that c-kit+ cells downregulate immune response in asthma by influencing local environment, possibly by cell-to-cell contact combined to paracrine action. In conclusion, intratracheally administered c-kit+ cells reduce inflammation, positively modulate airway remodeling, and improve function. These data document previously unrecognized properties of c-kit+ cells, able to impede pathophysiological features of experimental airway hyperresponsiveness.
Collapse
|
807
|
Engelmann C, Splith K, Berg T, Schmelzle M. Effects of granulocyte-colony stimulating factor (G-CSF) on stem cell mobilization in patients with liver failure. Eur J Intern Med 2016; 36:e37-e39. [PMID: 27745994 DOI: 10.1016/j.ejim.2016.09.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 09/07/2016] [Indexed: 12/23/2022]
Affiliation(s)
- Cornelius Engelmann
- University Hospital Leipzig, Section of Hepatology, Department of Internal Medicine, Neurology, Dermatology, Liebigstraße 20, 04103 Leipzig, Germany.
| | - Katrin Splith
- Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum, Department of Surgery, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Thomas Berg
- University Hospital Leipzig, Section of Hepatology, Department of Internal Medicine, Neurology, Dermatology, Liebigstraße 20, 04103 Leipzig, Germany.
| | - Moritz Schmelzle
- Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum, Department of Surgery, Augustenburger Platz 1, 13353 Berlin, Germany.
| |
Collapse
|
808
|
Han KH, Kim AK, Kim DI. Therapeutic Potential of Human Mesenchymal Stem Cells for Treating Ischemic Limb Diseases. Int J Stem Cells 2016; 9:163-168. [PMID: 27871151 PMCID: PMC5155711 DOI: 10.15283/ijsc16053] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2016] [Indexed: 12/19/2022] Open
Abstract
Ischemic limb diseases are induced by different obstructions of peripheral arteries. These obstructions result in insufficient nutrient and oxygen supplies to the extremities, thereby leading to severe tissue damage that is in turn related to severe morbidities and mortalities. Mesenchymal stem cells (MSCs) have been isolated from various sources. These cells are multipotent with respect to differentiation and are also characterized by migration, immune suppression, and secretion of paracrine factors. Mesenchymal stem cells have been proposed to have therapeutic potential for the treatment of ischemic limb diseases. In preclinical experiments, injection of single MSCs has been shown to increase angiogenesis and blood flow in ischemic hindlimb animal models; several molecular mechanisms of angiogenesis have also been elucidated. Furthermore, modified strategies have been developed for enhancing angiogenesis and the efficacy of MSCs. These strategies have demonstrated significant effects in pre-clinical studies. In clinical trials, MSCs have shown significant effects in the treatment of ischemic limb diseases. In this review, we focus on the therapeutic properties of human MSCs and the modified methods for enhancing angiogenesis in pre-clinical experiments. We also discuss the clinical applications of MSCs for treating limb ischemia.
Collapse
Affiliation(s)
- Kyu-Hyun Han
- Division of Vascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ae-Kyeong Kim
- Division of Vascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Dong-Ik Kim
- Division of Vascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
809
|
Ti D, Hao H, Fu X, Han W. Mesenchymal stem cells-derived exosomal microRNAs contribute to wound inflammation. SCIENCE CHINA-LIFE SCIENCES 2016; 59:1305-1312. [PMID: 27864711 DOI: 10.1007/s11427-016-0240-4] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 10/16/2016] [Indexed: 12/23/2022]
Abstract
Clinical and experimental studies have highlighted the significance of inflammation in coordinating wound repair and regeneration. However, it remains challenging to control the inflammatory response and tolerance at systemic levels without causing toxicity to injured tissues. Mesenchymal stem cells (MSCs) possess potent immunomodulatory properties and facilitate tissue repair by releasing exosomes, which generate a suitable microenvironment for inflammatory resolution. Exosomes contain several effective bioactive molecules and act as a cell-cell communication vehicle to influence cellular activities in recipient cells. During this process, the horizontal transfer of exosomal microRNAs (miRNAs) to acceptor cells, where they regulate target gene expression, is of particular interest for understanding the basic biology of inflammation ablation, tissue homeostasis, and development of therapeutic approaches. In this review, we describe a signature of three specific miRNAs (miR-21, miR-146a, and miR-181) present in human umbilical cord MSC-derived exosomes (MSC-EXO) identified microarray chip analysis and focus on the inflammatory regulatory functions of these immune-related miRNAs. We also discuss the potential mechanisms contributing to the resolution of wound inflammation and tissue healing.
Collapse
Affiliation(s)
- Dongdong Ti
- Institute of Basic Medicine, College of Life Sciences, Chinese PLA General Hospital, Beijing, 100853, China
| | - Haojie Hao
- Institute of Basic Medicine, College of Life Sciences, Chinese PLA General Hospital, Beijing, 100853, China
| | - Xiaobing Fu
- Institute of Basic Medicine, College of Life Sciences, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Weidong Han
- Institute of Basic Medicine, College of Life Sciences, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
810
|
Sherman LS, Shaker M, Mariotti V, Rameshwar P. Mesenchymal stromal/stem cells in drug therapy: New perspective. Cytotherapy 2016; 19:19-27. [PMID: 27765601 DOI: 10.1016/j.jcyt.2016.09.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 08/31/2016] [Accepted: 09/07/2016] [Indexed: 12/17/2022]
Abstract
Mesenchymal stromal/stem cells (MSC) have emerged as a class of cells suitable for cellular delivery of nanoparticles, drugs and micro-RNA cargo for targeted treatments such as tumor and other protective mechanisms. The special properties of MSC underscore the current use for various clinical applications. Examples of applications include but are not limited to regenerative medicine, immune disorders and anti-cancer therapies. In recent years, there has been intense research in modifying MSC to achieve targeted and efficient clinical outcomes. This review discusses effects of MSC in an inflammatory microenvironment and then explains how these properties could be important to the overall application of MSC in cell therapy. The article also advises caution in the application of these cells because of their role in tumorigenesis. The review stresses the use of MSC as vehicles for drug delivery and discusses the accompanying challenges, based on the influence of the microenvironment on MSC.
Collapse
Affiliation(s)
- Lauren S Sherman
- Graduate School of Biomedical Sciences, Division of Hematology/Oncology, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA; Department of Medicine, Division of Hematology/Oncology, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Maran Shaker
- Graduate School of Biomedical Sciences, Division of Hematology/Oncology, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Veronica Mariotti
- Department of Medicine, Division of Hematology/Oncology, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Pranela Rameshwar
- Graduate School of Biomedical Sciences, Division of Hematology/Oncology, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA; Department of Medicine, Division of Hematology/Oncology, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA.
| |
Collapse
|
811
|
Preconditioning of Human Mesenchymal Stem Cells to Enhance Their Regulation of the Immune Response. Stem Cells Int 2016; 2016:3924858. [PMID: 27822228 PMCID: PMC5086389 DOI: 10.1155/2016/3924858] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 09/28/2016] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have attracted the attention of researchers and clinicians for their ability to differentiate into a number of cell types, participate in tissue regeneration, and repair the damaged tissues by producing various growth factors and cytokines, as well as their unique immunoprivilege in alloreactive hosts. The immunomodulatory functions of exogenous MSCs have been widely investigated in immune-mediated inflammatory diseases and transplantation research. However, a harsh environment at the site of tissue injury/inflammation with insufficient oxygen supply, abundance of reactive oxygen species, and presence of other harmful molecules that damage the adoptively transferred cells collectively lead to low survival and engraftment of the transferred cells. Preconditioning of MSCs ex vivo by hypoxia, inflammatory stimulus, or other factors/conditions prior to their use in therapy is an adaptive strategy that prepares MSCs to survive in the harsh environment and to enhance their regulatory function of the local immune responses. This review focuses on a number of approaches in preconditioning human MSCs with the goal of augmenting their capacity to regulate both innate and adaptive immune responses.
Collapse
|
812
|
Saparov A, Ogay V, Nurgozhin T, Jumabay M, Chen WCW. Preconditioning of Human Mesenchymal Stem Cells to Enhance Their Regulation of the Immune Response. Stem Cells Int 2016; 2016:3924858. [PMID: 27822228 PMCID: PMC5086389 DOI: 10.1155/2016/3924858 10.1155/2016/3924858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 09/28/2016] [Indexed: 03/24/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have attracted the attention of researchers and clinicians for their ability to differentiate into a number of cell types, participate in tissue regeneration, and repair the damaged tissues by producing various growth factors and cytokines, as well as their unique immunoprivilege in alloreactive hosts. The immunomodulatory functions of exogenous MSCs have been widely investigated in immune-mediated inflammatory diseases and transplantation research. However, a harsh environment at the site of tissue injury/inflammation with insufficient oxygen supply, abundance of reactive oxygen species, and presence of other harmful molecules that damage the adoptively transferred cells collectively lead to low survival and engraftment of the transferred cells. Preconditioning of MSCs ex vivo by hypoxia, inflammatory stimulus, or other factors/conditions prior to their use in therapy is an adaptive strategy that prepares MSCs to survive in the harsh environment and to enhance their regulatory function of the local immune responses. This review focuses on a number of approaches in preconditioning human MSCs with the goal of augmenting their capacity to regulate both innate and adaptive immune responses.
Collapse
Affiliation(s)
- Arman Saparov
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana 010000, Kazakhstan
| | - Vyacheslav Ogay
- Stem Cell Laboratory, National Center for Biotechnology, Astana 010000, Kazakhstan
| | - Talgat Nurgozhin
- Center for Life Sciences, Nazarbayev University, Astana 010000, Kazakhstan
| | - Medet Jumabay
- Division of Cardiology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - William C. W. Chen
- Research Laboratory of Electronics and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
813
|
Matula Z, Németh A, Lőrincz P, Szepesi Á, Brózik A, Buzás EI, Lőw P, Német K, Uher F, Urbán VS. The Role of Extracellular Vesicle and Tunneling Nanotube-Mediated Intercellular Cross-Talk Between Mesenchymal Stem Cells and Human Peripheral T Cells. Stem Cells Dev 2016; 25:1818-1832. [PMID: 27596268 DOI: 10.1089/scd.2016.0086] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The role of extracellular vesicles (EVs) in mediating the immunosuppressory properties of mesenchymal stem cells (MSCs) has recently attracted remarkable scientific interest. The aim of this work was to analyze the transport mechanisms of membrane and cytoplasmic components between T lymphocytes and adipose tissue-derived MSCs (AD-MSCs), by focusing on the role of distinct populations of EVs, direct cell-cell contacts, and the soluble mediators per se in modulating T lymphocyte function. We found that neither murine thymocytes and human primary T cells nor Jurkat lymphoblastoid cells incorporated appreciable amounts of MSC-derived microvesicles (MVs) or exosomes (EXOs). Moreover, these particles had no effect on the proliferation and IFN-γ production of in vitro-stimulated primary T cells. In contrast, AD-MSCs incorporated large amounts of membrane components from T cells as an intensive uptake of EXOs and MVs could be observed. Interestingly, we found a bidirectional exchange of cytoplasmic components between human AD-MSCs and primary T lymphocytes, mediated by tunneling nanotubes (TNTs) derived exclusively from the T cells. In contrast, TNTs couldn't be observed between AD-MSCs and the Jurkat cells. Our results reveal a novel and efficient way of intercellular communication between MSCs and T cells, and may help a better understanding of the immunomodulatory function of MSCs.
Collapse
Affiliation(s)
- Zsolt Matula
- 1 Institute of Enzymology , Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Andrea Németh
- 2 Department of Genetics, Cell- and Immunobiology, Semmelweis University , Budapest, Hungary
| | - Péter Lőrincz
- 3 Department of Anatomy, Cell and Developmental Biology, Eötvos Loránd University , Budapest, Hungary
| | - Áron Szepesi
- 1 Institute of Enzymology , Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Anna Brózik
- 1 Institute of Enzymology , Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Edit Irén Buzás
- 2 Department of Genetics, Cell- and Immunobiology, Semmelweis University , Budapest, Hungary
| | - Péter Lőw
- 3 Department of Anatomy, Cell and Developmental Biology, Eötvos Loránd University , Budapest, Hungary
| | | | - Ferenc Uher
- 5 Stem Cell Biology, National Blood Service , Budapest, Hungary
| | - Veronika S Urbán
- 6 Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University , Budapest, Hungary
| |
Collapse
|
814
|
Maumus M, Roussignol G, Toupet K, Penarier G, Bentz I, Teixeira S, Oustric D, Jung M, Lepage O, Steinberg R, Jorgensen C, Noel D. Utility of a Mouse Model of Osteoarthritis to Demonstrate Cartilage Protection by IFNγ-Primed Equine Mesenchymal Stem Cells. Front Immunol 2016; 7:392. [PMID: 27729913 PMCID: PMC5037129 DOI: 10.3389/fimmu.2016.00392] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 09/15/2016] [Indexed: 12/30/2022] Open
Abstract
Objective Mesenchymal stem cells isolated from adipose tissue (ASC) have been shown to influence the course of osteoarthritis (OA) in different animal models and are promising in veterinary medicine for horses involved in competitive sport. The aim of this study was to characterize equine ASCs (eASCs) and investigate the role of interferon-gamma (IFNγ)-priming on their therapeutic effect in a murine model of OA, which could be relevant to equine OA. Methods ASC were isolated from subcutaneous fat. Expression of specific markers was tested by cytometry and RT-qPCR. Differentiation potential was evaluated by histology and RT-qPCR. For functional assays, naïve or IFNγ-primed eASCs were cocultured with peripheral blood mononuclear cells or articular cartilage explants. Finally, the therapeutic effect of eASCs was tested in the model of collagenase-induced OA (CIOA) in mice. Results The immunosuppressive function of eASCs on equine T cell proliferation and their chondroprotective effect on equine cartilage explants were demonstrated in vitro. Both cartilage degradation and T cell activation were reduced by naïve and IFNγ-primed eASCs, but IFNγ-priming enhanced these functions. In CIOA, intra-articular injection of eASCs prevented articular cartilage from degradation and IFNγ-primed eASCs were more potent than naïve cells. This effect was related to the modulation of eASC secretome by IFNγ-priming. Conclusion IFNγ-priming of eASCs potentiated their antiproliferative and chondroprotective functions. We demonstrated that the immunocompetent mouse model of CIOA was relevant to test the therapeutic efficacy of xenogeneic eASCs for OA and confirmed that IFNγ-primed eASCs may have a therapeutic value for musculoskeletal diseases in veterinary medicine.
Collapse
Affiliation(s)
- Marie Maumus
- U1183, INSERM, Hôpital Saint-Eloi, Montpellier, France; Montpellier University, UFR de Médecine, Montpellier, France
| | | | - Karine Toupet
- U1183, INSERM, Hôpital Saint-Eloi, Montpellier, France; Montpellier University, UFR de Médecine, Montpellier, France
| | | | | | | | | | | | - Olivier Lepage
- GREMERES-ICE, University of Lyon , Marcy l'Etoile , France
| | | | - Christian Jorgensen
- U1183, INSERM, Hôpital Saint-Eloi, Montpellier, France; Montpellier University, UFR de Médecine, Montpellier, France; Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Hôpital Lapeyronie, Montpellier, France
| | - Danièle Noel
- U1183, INSERM, Hôpital Saint-Eloi, Montpellier, France; Montpellier University, UFR de Médecine, Montpellier, France; Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Hôpital Lapeyronie, Montpellier, France
| |
Collapse
|
815
|
Dynamic Tracking Human Mesenchymal Stem Cells Tropism following Smoke Inhalation Injury in NOD/SCID Mice. Stem Cells Int 2016; 2016:1691856. [PMID: 27725837 PMCID: PMC5048056 DOI: 10.1155/2016/1691856] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 06/22/2016] [Accepted: 06/28/2016] [Indexed: 12/12/2022] Open
Abstract
Multiple preclinical evidences have supported the potential value of mesenchymal stem cells (MSCs) for treatment of acute lung injury (ALI). However, few studies focus on the dynamic tropism of MSCs in animals with acute lung injury. In this study, we track systemically transplanted human bone marrow-derived mesenchymal stem cells (hBMSCs) in NOD/SCID mice with smoke inhalation injury (SII) through bioluminescence imaging (BLI). The results showed that hBMSCs systemically delivered into healthy NOD/SCID mouse initially reside in the lungs and then partially translocate to the abdomen after 24 h. Compared with the uninjured control group treated with hBMSCs, higher numbers of hBMSCs were found in the lungs of the SII NOD/SCID mice. In both the uninjured and SII mice, the BLI signals in the lungs steadily decreased over time and disappeared by 5 days after treatment. hBMSCs significantly attenuated lung injury, elevated the levels of KGF, decreased the levels of TNF-α in BALF, and inhibited inflammatory cell infiltration in the mice with SII. In conclusion, our findings demonstrated that more systemically infused hBMSCs localized to the lungs in mice with SII. hBMSC xenografts repaired smoke inhalation-induced lung injury in mice. This repair was maybe due to the effect of anti-inflammatory and secreting KGF of hMSCs but not associated with the differentiation of the hBMSCs into alveolar epithelial cells.
Collapse
|
816
|
Zhang Y, Yu Z, Jiang D, Liang X, Liao S, Zhang Z, Yue W, Li X, Chiu SM, Chai YH, Liang Y, Chow Y, Han S, Xu A, Tse HF, Lian Q. iPSC-MSCs with High Intrinsic MIRO1 and Sensitivity to TNF-α Yield Efficacious Mitochondrial Transfer to Rescue Anthracycline-Induced Cardiomyopathy. Stem Cell Reports 2016; 7:749-763. [PMID: 27641650 PMCID: PMC5063626 DOI: 10.1016/j.stemcr.2016.08.009] [Citation(s) in RCA: 195] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 08/11/2016] [Accepted: 08/16/2016] [Indexed: 12/31/2022] Open
Abstract
Mesenchymal stem cells (MSCs) can donate mitochondria and rescue anthracycline-induced cardiomyocyte (CM) damage, although the underlying mechanisms remain elusive. We determined that the superior efficiency of mitochondrial transfer by human induced-pluripotent-stem-cell-derived MSCs (iPSC-MSCs) compared with bone marrow-derived MSCs (BM-MSCs) is due to high expression of intrinsic Rho GTPase 1 (MIRO1). Further, due to a higher level of TNFαIP2 expression, iPSC-MSCs are more responsive to tumor necrosis factor alpha (TNF-α)-induced tunneling nanotube (TNT) formation for mitochondrial transfer to CMs, which is regulated via the TNF-α/NF-κB/TNFαIP2 signaling pathway. Inhibition of TNFαIP2 or MIRO1 in iPSC-MSCs reduced the efficiency of mitochondrial transfer and decreased CMs protection. Compared with BM-MSCs, transplantation of iPSC-MSCs into a mouse model of anthracycline-induced cardiomyopathy resulted in more human mitochondrial retention and bioenergetic preservation in heart tissue. Efficacious transfer of mitochondria from iPSC-MSCs to CMs, due to higher MIRO1 expression and responsiveness to TNF-α-induced nanotube formation, effectively attenuates anthracycline-induced CM damage. Functional mitochondrial transfer of iPSC-MSCs attenuates Dox-induced cardiomyopathy High intrinsic Miro1 in iPSC-MSCs contributes to efficacious mitochondrial transfer iPSC-MSCs are highly responsive to TNF-α-induced tunneling nanotube formation
Collapse
Affiliation(s)
- Yuelin Zhang
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Zhendong Yu
- Central Laboratory, Peking University Shenzhen Hospital, Shenzhen 518000, China
| | - Dan Jiang
- Department of Ophthalmology, The University of Hong Kong, Hong Kong, China
| | - Xiaoting Liang
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Songyan Liao
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Zhao Zhang
- Department of Ophthalmology, The University of Hong Kong, Hong Kong, China
| | - Wensheng Yue
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xiang Li
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Sin-Ming Chiu
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yuet-Hung Chai
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong, China; Research Centre of Heart, Brain, Hormone, and Healthy Aging, The University of Hong Kong, Hong Kong, China
| | - Yingmin Liang
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yenyen Chow
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Shuo Han
- Department of Ophthalmology, The University of Hong Kong, Hong Kong, China
| | - Aimin Xu
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong, China; Research Centre of Heart, Brain, Hormone, and Healthy Aging, The University of Hong Kong, Hong Kong, China
| | - Hung-Fat Tse
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong, China; Research Centre of Heart, Brain, Hormone, and Healthy Aging, The University of Hong Kong, Hong Kong, China; Hong Kong-Guangdong Joint Laboratory on Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, The University of Hong Kong, Hong Kong, China.
| | - Qizhou Lian
- Cardiology Division, Department of Medicine, The University of Hong Kong, Hong Kong, China; Department of Ophthalmology, The University of Hong Kong, Hong Kong, China; Research Centre of Heart, Brain, Hormone, and Healthy Aging, The University of Hong Kong, Hong Kong, China; Shenzhen Institutes of Research and Innovation, The University of Hong Kong, Shenzhen 518000, China.
| |
Collapse
|
817
|
Irvine SA, Venkatraman SS. Bioprinting and Differentiation of Stem Cells. Molecules 2016; 21:E1188. [PMID: 27617991 PMCID: PMC6273261 DOI: 10.3390/molecules21091188] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 08/26/2016] [Accepted: 08/26/2016] [Indexed: 01/10/2023] Open
Abstract
The 3D bioprinting of stem cells directly into scaffolds offers great potential for the development of regenerative therapies; in particular for the fabrication of organ and tissue substitutes. For this to be achieved; the lineage fate of bioprinted stem cell must be controllable. Bioprinting can be neutral; allowing culture conditions to trigger differentiation or alternatively; the technique can be designed to be stimulatory. Such factors as the particular bioprinting technique; bioink polymers; polymer cross-linking mechanism; bioink additives; and mechanical properties are considered. In addition; it is discussed that the stimulation of stem cell differentiation by bioprinting may lead to the remodeling and modification of the scaffold over time matching the concept of 4D bioprinting. The ability to tune bioprinting properties as an approach to fabricate stem cell bearing scaffolds and to also harness the benefits of the cells multipotency is of considerable relevance to the field of biomaterials and bioengineering.
Collapse
Affiliation(s)
- Scott A Irvine
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore.
| | - Subbu S Venkatraman
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore.
| |
Collapse
|
818
|
Cao J, Hou S, Ding H, Liu Z, Song M, Qin X, Wang X, Yu M, Sun Z, Liu J, Sun S, Xiao P, Lv Q, Fan H. In Vivo Tracking of Systemically Administered Allogeneic Bone Marrow Mesenchymal Stem Cells in Normal Rats through Bioluminescence Imaging. Stem Cells Int 2016; 2016:3970942. [PMID: 27610137 PMCID: PMC5005574 DOI: 10.1155/2016/3970942] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 05/26/2016] [Accepted: 07/03/2016] [Indexed: 01/14/2023] Open
Abstract
Recently, mesenchymal stem cells (MSCs) are increasingly used as a panacea for multiple types of disease short of effective treatment. Dozens of clinical trials published demonstrated strikingly positive therapeutic effects of MSCs. However, as a specific agent, little research has focused on the dynamic distribution of MSCs after in vivo administration. In this study, we track systemically transplanted allogeneic bone marrow mesenchymal stem cells (BMSCs) in normal rats through bioluminescence imaging (BLI) in real time. Ex vivo organ imaging, immunohistochemistry (IHC), and RT-PCR were conducted to verify the histological distribution of BMSCs. Our results showed that BMSCs home to the dorsal skin apart from the lungs and kidneys after tail vein injection and could not be detected 14 days later. Allogeneic BMSCs mainly appeared not at the parenchymatous organs but at the subepidermal connective tissue and adipose tissue in healthy rats. There were no significant MSCs-related adverse effects except for transient decrease in neutrophils. These findings will provide experimental evidences for a better understanding of the biocharacteristics of BMSCs.
Collapse
Affiliation(s)
- Juan Cao
- Institute of Disaster Medicine and Public Health, Affiliated Hospital of Logistic University of Chinese People's Armed Police Force, Tianjin, China
- Key Laboratory of Emergency and Disaster Medicine in Chinese People's Liberation Army (PLA), Tianjin 300162, China
| | - Shike Hou
- Institute of Disaster Medicine and Public Health, Affiliated Hospital of Logistic University of Chinese People's Armed Police Force, Tianjin, China
- Key Laboratory of Emergency and Disaster Medicine in Chinese People's Liberation Army (PLA), Tianjin 300162, China
| | - Hui Ding
- Institute of Disaster Medicine and Public Health, Affiliated Hospital of Logistic University of Chinese People's Armed Police Force, Tianjin, China
- Key Laboratory of Emergency and Disaster Medicine in Chinese People's Liberation Army (PLA), Tianjin 300162, China
| | - Ziquan Liu
- Institute of Disaster Medicine and Public Health, Affiliated Hospital of Logistic University of Chinese People's Armed Police Force, Tianjin, China
- Key Laboratory of Emergency and Disaster Medicine in Chinese People's Liberation Army (PLA), Tianjin 300162, China
| | - Meijuan Song
- Institute of Disaster Medicine and Public Health, Affiliated Hospital of Logistic University of Chinese People's Armed Police Force, Tianjin, China
- Key Laboratory of Emergency and Disaster Medicine in Chinese People's Liberation Army (PLA), Tianjin 300162, China
| | - Xiaojing Qin
- Department of Pathology, Affiliated Hospital of Logistic University of Chinese People's Armed Police Force, Tianjin, China
| | - Xue Wang
- Institute of Disaster Medicine and Public Health, Affiliated Hospital of Logistic University of Chinese People's Armed Police Force, Tianjin, China
- Key Laboratory of Emergency and Disaster Medicine in Chinese People's Liberation Army (PLA), Tianjin 300162, China
| | - Mengyang Yu
- Institute of Disaster Medicine and Public Health, Affiliated Hospital of Logistic University of Chinese People's Armed Police Force, Tianjin, China
- Key Laboratory of Emergency and Disaster Medicine in Chinese People's Liberation Army (PLA), Tianjin 300162, China
| | - Zhiguang Sun
- Institute of Disaster Medicine and Public Health, Affiliated Hospital of Logistic University of Chinese People's Armed Police Force, Tianjin, China
- Key Laboratory of Emergency and Disaster Medicine in Chinese People's Liberation Army (PLA), Tianjin 300162, China
| | - Jinyang Liu
- Institute of Disaster Medicine and Public Health, Affiliated Hospital of Logistic University of Chinese People's Armed Police Force, Tianjin, China
- Key Laboratory of Emergency and Disaster Medicine in Chinese People's Liberation Army (PLA), Tianjin 300162, China
| | - Shuli Sun
- Institute of Disaster Medicine and Public Health, Affiliated Hospital of Logistic University of Chinese People's Armed Police Force, Tianjin, China
- Key Laboratory of Emergency and Disaster Medicine in Chinese People's Liberation Army (PLA), Tianjin 300162, China
| | - Peixin Xiao
- Institute of Disaster Medicine and Public Health, Affiliated Hospital of Logistic University of Chinese People's Armed Police Force, Tianjin, China
- Key Laboratory of Emergency and Disaster Medicine in Chinese People's Liberation Army (PLA), Tianjin 300162, China
| | - Qi Lv
- Institute of Disaster Medicine and Public Health, Affiliated Hospital of Logistic University of Chinese People's Armed Police Force, Tianjin, China
- Key Laboratory of Emergency and Disaster Medicine in Chinese People's Liberation Army (PLA), Tianjin 300162, China
| | - Haojun Fan
- Institute of Disaster Medicine and Public Health, Affiliated Hospital of Logistic University of Chinese People's Armed Police Force, Tianjin, China
- Key Laboratory of Emergency and Disaster Medicine in Chinese People's Liberation Army (PLA), Tianjin 300162, China
| |
Collapse
|
819
|
Mesenchymal Stem Cells after Polytrauma: Actor and Target. Stem Cells Int 2016; 2016:6289825. [PMID: 27340408 PMCID: PMC4909902 DOI: 10.1155/2016/6289825] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 05/09/2016] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells that are considered indispensable in regeneration processes after tissue trauma. MSCs are recruited to damaged areas via several chemoattractant pathways where they function as “actors” in the healing process by the secretion of manifold pro- and anti-inflammatory, antimicrobial, pro- and anticoagulatory, and trophic/angiogenic factors, but also by proliferation and differentiation into the required cells. On the other hand, MSCs represent “targets” during the pathophysiological conditions after severe trauma, when excessively generated inflammatory mediators, complement activation factors, and damage- and pathogen-associated molecular patterns challenge MSCs and alter their functionality. This in turn leads to complement opsonization, lysis, clearance by macrophages, and reduced migratory and regenerative abilities which culminate in impaired tissue repair. We summarize relevant cellular and signaling mechanisms and provide an up-to-date overview about promising future therapeutic MSC strategies in the context of severe tissue trauma.
Collapse
|