651
|
Assessment of the Immunosuppressive Potential of INF-γ Licensed Adipose Mesenchymal Stem Cells, Their Secretome and Extracellular Vesicles. Cells 2019; 8:cells8010022. [PMID: 30621275 PMCID: PMC6356584 DOI: 10.3390/cells8010022] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 12/25/2018] [Accepted: 12/29/2018] [Indexed: 12/11/2022] Open
Abstract
There is an active search for the ideal strategy to potentialize the effects of Mesenchymal Stem-Cells (MSCs) over the immune system. Also, part of the scientific community is seeking to elucidate the therapeutic potential of MSCs secretome and its extracellular vesicles (EVs), in order to avoid the complexity of a cellular therapy. Here, we investigate the effects of human adipose MSCs (AMSCs) licensing with INF-γ and TLR3 agonist over AMSCs proliferation, migration, as well as the immunomodulatory function. Furthermore, we evaluated how the licensing of AMSCs affected the immunomodulatory function of AMSC derived-secretome, including their EVs. INF-γ licensed-AMSCs presented an elevated expression of indoleamine 2,3-dioxygenase (IDO), accompanied by increased ICAM-1, as well as a higher immunosuppressive potential, compared to unlicensed AMSCs. Interestingly, the conditioned medium obtained from INF-γ licensed-AMSCs also revealed a slightly superior immunosuppressive potential, compared to other licensing strategies. Therefore, unlicensed and INF-γ licensed-AMSCs groups were used to isolate EVs. Interestingly, EVs isolated from both groups displayed similar capacity to inhibit T-cell proliferation. EVs isolated from both groups shared similar TGF-β and Galectin-1 mRNA content but only EVs derived from INF-γ licensed-AMSCs expressed IDO mRNA. In summary, we demonstrated that INF-γ licensing of AMSCs provides an immunosuppressive advantage both from a cell-cell contact-dependent perspective, as well as in a cell-free context. Interestingly, EVs derived from unlicensed and INF-γ licensed-AMSCs have similar ability to control activated T-cell proliferation. These results contribute towards the development of new strategies to control the immune response based on AMSCs or their derived products.
Collapse
|
652
|
Al-Sowayan B, Keogh RJ, Abumaree M, Georgiou HM, Kalionis B. An ex vivo human placental vessel perfusion method to study mesenchymal stem/stromal cell migration. Stem Cell Investig 2019; 6:2. [PMID: 30842969 DOI: 10.21037/sci.2018.12.03] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 12/11/2018] [Indexed: 12/28/2022]
Abstract
Background To initiate tissue repair, mesenchymal stem/stromal cells (MSCs) must enter the blood stream, migrate to the targeted area, cross the endothelial barrier and home to the damaged tissue. This process is not yet fully understood in humans and thus, the aim of this study was to develop an ex vivo placental vessel perfusion method to examine human MSC movement from a blood vessel into human tissue. This will provide a better understanding of MSC migration, movement through the endothelial barrier and engraftment into target tissue, in a setting that more closely represents the in vivo state, compared with conventional in vitro human cell culture models. Moreover, important similarities and differences to animal experimental model systems may be revealed by this method. Methods Human placental hTERT transformed MSC lines were labelled with live-cell fluorescence dyes, and then perfused into term human placental blood vessel. After labelled MSCs were perfused into the vessel, the vessel was dissected from the placenta and incubated at cell growth conditions. Following incubation, the vessel was washed thoroughly to remove unattached, labelled MSCs and then snap frozen for sectioning. After sectioning, immunofluorescence staining of the endothelium was carried out to detect if labelled MSCs crossed the endothelial barrier. Results Twelve placental vessel perfusions were successfully completed. In eight of the twelve perfused vessels, qualitative assessment of immunofluorescence in sections (n=20, 5 µm sections/vessel) revealed labelled MSCs had crossed the endothelial barrier. Conclusions The human placental ex vivo vessel perfusion method could be used to assess human MSC migration into human tissue. Cells of the MSC lines were able to adhere and transmigrate through the endothelial barrier in a manner similar to that of leukocytes. Notably, cells that transmigrated remained in close proximity to the endothelium, which is consistent with the reported MSC vascular niche in placental blood vessels.
Collapse
Affiliation(s)
- Balta Al-Sowayan
- Pregnancy Research Centre, Department of Maternal-Fetal Medicine, Royal Women's Hospital, Parkville, Victoria 3052, Australia.,University of Melbourne Department of Obstetrics and Gynaecology, Royal Women's Hospital, Parkville, Victoria 3052, Australia.,Stem Cell and Regenerative Medicine Department, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh 11426, Saudi Arabia
| | - Rosemary J Keogh
- Pregnancy Research Centre, Department of Maternal-Fetal Medicine, Royal Women's Hospital, Parkville, Victoria 3052, Australia.,University of Melbourne Department of Obstetrics and Gynaecology, Royal Women's Hospital, Parkville, Victoria 3052, Australia
| | - Mohammed Abumaree
- Stem Cell and Regenerative Medicine Department, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh 11426, Saudi Arabia.,College of Science and Health Professions, King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh 11481, Saudi Arabia
| | - Harry M Georgiou
- Pregnancy Research Centre, Department of Maternal-Fetal Medicine, Royal Women's Hospital, Parkville, Victoria 3052, Australia.,University of Melbourne Department of Obstetrics and Gynaecology, Royal Women's Hospital, Parkville, Victoria 3052, Australia
| | - Bill Kalionis
- Pregnancy Research Centre, Department of Maternal-Fetal Medicine, Royal Women's Hospital, Parkville, Victoria 3052, Australia.,University of Melbourne Department of Obstetrics and Gynaecology, Royal Women's Hospital, Parkville, Victoria 3052, Australia
| |
Collapse
|
653
|
Razmkhah M, Abtahi S, Ghaderi A. Mesenchymal Stem Cells, Immune Cells and Tumor Cells Crosstalk: A Sinister Triangle in the Tumor Microenvironment. Curr Stem Cell Res Ther 2019; 14:43-51. [PMID: 30112998 DOI: 10.2174/1574888x13666180816114809] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 08/08/2018] [Accepted: 08/09/2018] [Indexed: 02/07/2023]
Abstract
Mesenchymal Stem Cells [MSCs] are a heterogeneous population of fibroblast-like cells which maintain self-renewability and pluripotency. Many studies have demonstrated the immunomodulatory effects of MSCs on the innate and adaptive immune cells. As a result of interactions with tumor cells, microenvironment and immune-stimulating milieu, MSCs contribute to tumor progression by several mechanisms, including sustained proliferative signal in cancer stem cells [CSCs], inhibition of tumor cell apoptosis, transition to tumor-associated fibroblasts [TAFs], promotion of angiogenesis, stimulation of epithelial-mesenchymal transition [EMT], suppression of immune responses, and consequential promotion of tumor metastasis. Here, we present an overview of the latest findings on Janusfaced roles that MSCs play in the tumor microenvironment [TME], with a concise focus on innate and adaptive immune responses.
Collapse
Affiliation(s)
- Mahboobeh Razmkhah
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shabnam Abtahi
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abbas Ghaderi
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
654
|
Myneni VD, McClain-Caldwell I, Martin D, Vitale-Cross L, Marko K, Firriolo JM, Labow BI, Mezey E. Mesenchymal stromal cells from infants with simple polydactyly modulate immune responses more efficiently than adult mesenchymal stromal cells. Cytotherapy 2018; 21:148-161. [PMID: 30595353 DOI: 10.1016/j.jcyt.2018.11.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 11/04/2018] [Accepted: 11/29/2018] [Indexed: 12/23/2022]
Abstract
Bone marrow-derived stromal cells or mesenchymal stromal cells (BMSCs or MSCs, as we will call them in this work) are multipotent progenitor cells that can differentiate into osteoblasts, adipocytes and chondrocytes. In addition, MSCs have been shown to modulate the function of a variety of immune cells. Donor age has been shown to affect the regenerative potential, differentiation, proliferation and anti-inflammatory potency of MSCs; however, the impact of donor age on their immunosuppressive activity is unknown. In this study, we evaluated the ability of MSCs derived from very young children and adults on T-cell suppression and cytokine secretion by monocytes/macrophages. MSCs were obtained from extra digits of children between 10 and 21 months and adults between 28 and 64 years of age. We studied cell surface marker expression, doubling time, lineage differentiation potential and immunosuppressive function of the MSCs. Young MSCs double more quickly and differentiate into bone and fat cells more efficiently than those from older donors. They also form more and dense colonies of fibroblasts (colony forming unit-fibroblast [CFU-F]). MSCs from both young and adult subjects suppressed T-cell proliferation in a mitogen-induced assay at 1:3 and 1:30 ratios. At a 1:30 ratio, however, MSCs from adults did not, but MSCs from infants did suppress T-cell proliferation. In the mixed lymphocyte reaction assay, MSCs from infants produced similar levels of suppression at all three MSC/T-cell ratios, but adult MSCs only inhibited T-cell proliferation at a 1:3 ratio. Cytokine analyses of co-cultures of MSCs and macrophages showed that both adult and young MSCs suppress tumor necrosis factor alpha (TNF-α) and induce interleukin-10 (IL-10) production in macrophage co-culture assay in a similar manner. Overall, this work shows that developing MSCs display a higher level of immunosuppression than mature MSCs.
Collapse
Affiliation(s)
- Vamsee D Myneni
- Adult Stem Cell Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Ian McClain-Caldwell
- Adult Stem Cell Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Daniel Martin
- Genomics & Computational Biology Core, National Institute of Dental and Craniofacial Research, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, USA
| | - Lynn Vitale-Cross
- Adult Stem Cell Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Karoly Marko
- Adult Stem Cell Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Joseph M Firriolo
- Department of Plastic and Oral Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Brian I Labow
- Department of Plastic and Oral Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Eva Mezey
- Adult Stem Cell Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
655
|
Centeno C, Sheinkop M, Dodson E, Stemper I, Williams C, Hyzy M, Ichim T, Freeman M. A specific protocol of autologous bone marrow concentrate and platelet products versus exercise therapy for symptomatic knee osteoarthritis: a randomized controlled trial with 2 year follow-up. J Transl Med 2018; 16:355. [PMID: 30545387 PMCID: PMC6293635 DOI: 10.1186/s12967-018-1736-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 12/06/2018] [Indexed: 01/06/2023] Open
Abstract
Background Cell-based therapies have shown promise for the treatment of knee osteoarthritis (OA). The current study compared exercise therapy to autologous bone marrow concentrate (BMC) and platelet products for knee OA treatment. Methods Patients with symptomatic knee OA (N = 48) were randomized into either an exercise therapy control group or treatment group with injection of autologous BMC and platelet products. Patients in the control group could crossover to BMC treatment after 3 months. Clinical outcomes were documented at baseline and at 6-weeks, 3, 6, 12 and 24 months, including the Knee Society Score (KSS), Pain Visual Analogue Scale, Short Form-12 Scales (SF-12), and Lower Extremity Activity Scale (LEAS). Results All patients in the exercise group crossed over to receive BMC treatment after 3 months (N = 22 crossover). At 3 months, KSS-knee, SF-12 Physical, and LEAS improved significantly in the crossover group compared to exercise, similar to significant improvements on KSS-knee and LEAS for the treatment group (N = 26) compared to exercise group at 3 months. After BMC treatment, patients’ clinical outcome scores (except SF-12 Mental Health), were significantly improved through the 2-year follow-up compared to baseline. No serious adverse events were reported. Conclusion The use of image-guided percutaneous BMC with platelet products yielded better results than exercise therapy as an effective alternative therapy for patients with symptomatic moderate to moderate-severe osteoarthritis of the knee. Trial registration NCT02034032. https://clinicaltrials.gov/ct2/show/NCT02034032. Registered 13 January 2014
Collapse
Affiliation(s)
- Christopher Centeno
- Centeno-Schultz Clinic, 403 Summit Blvd Suite 201, Broomfield, CO, 80021, USA.,Regenexx, LLC, Des Moines, IA, USA
| | | | | | | | | | - Matthew Hyzy
- Centeno-Schultz Clinic, 403 Summit Blvd Suite 201, Broomfield, CO, 80021, USA
| | | | - Michael Freeman
- CAPHRI, School of Public Health and Primary Care, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
656
|
Mitochondrial Neuroglobin Is Necessary for Protection Induced by Conditioned Medium from Human Adipose-Derived Mesenchymal Stem Cells in Astrocytic Cells Subjected to Scratch and Metabolic Injury. Mol Neurobiol 2018; 56:5167-5187. [PMID: 30536184 DOI: 10.1007/s12035-018-1442-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 11/29/2018] [Indexed: 12/27/2022]
Abstract
Astrocytes are specialized cells capable of regulating inflammatory responses in neurodegenerative diseases or traumatic brain injury. In addition to playing an important role in neuroinflammation, these cells regulate essential functions for the preservation of brain tissue. Therefore, the search for therapeutic alternatives to preserve these cells and maintain their functions contributes in some way to counteract the progress of the injury and maintain neuronal survival in various brain pathologies. Among these strategies, the conditioned medium from human adipose-derived mesenchymal stem cells (CM-hMSCA) has been reported with a potential beneficial effect against several neuropathologies. In this study, we evaluated the potential effect of CM-hMSCA in a model of human astrocytes (T98G cells) subjected to scratch injury. Our findings demonstrated that CM-hMSCA regulates the cytokines IL-2, IL-6, IL-8, IL-10, GM-CSF, and TNF-α, downregulates calcium at the cytoplasmic level, and regulates mitochondrial dynamics and the respiratory chain. These actions are accompanied by modulation of the expression of different proteins involved in signaling pathways such as AKT/pAKT and ERK1/2/pERK, and may mediate the localization of neuroglobin (Ngb) at the cellular level. We also confirmed that Ngb mediated the protective effects of CM-hMSCA through regulation of proteins involved in survival pathways and oxidative stress. In conclusion, regulation of brain inflammation combined with the recovery of fundamental cellular aspects in the face of injury makes CM-hMSCA a promising candidate for the protection of astrocytes in brain pathologies.
Collapse
|
657
|
Robb KP, Fitzgerald JC, Barry F, Viswanathan S. Mesenchymal stromal cell therapy: progress in manufacturing and assessments of potency. Cytotherapy 2018; 21:289-306. [PMID: 30528726 DOI: 10.1016/j.jcyt.2018.10.014] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 10/26/2018] [Accepted: 10/26/2018] [Indexed: 02/06/2023]
Abstract
Mesenchymal stromal cell (MSC) therapies have been pursued for a broad spectrum of indications but mixed reports on clinical efficacy have given rise to some degree of skepticism regarding the effectiveness of this approach. However, recent reports of successful clinical outcomes and regulatory approvals for graft-versus-host disease, Crohn's disease and critical limb ischemia have prompted a shift in this perspective. With hundreds of clinical trials involving MSCs currently underway and an increasing demand for large-scale manufacturing protocols, there is a critical need to develop standards that can be applied to processing methods and to establish consensus assays for both MSC processing control and MSC product release. Reference materials and validated, uniformly applied tests for quality control of MSC products are needed. Here, we review recent developments in MSC manufacturing technologies, release testing and potency assays. We conclude that, although MSCs hold considerable promise clinically, economies of scale have yet to be achieved although numerous bioreactor technologies for scalable production of MSCs exist. Additionally, rigorous disease-specific product testing and comprehensive understanding of mechanisms of action, which are linked to relevant process and product release potency assays, will be required to ensure that these therapies continue to be successful.
Collapse
Affiliation(s)
- Kevin P Robb
- The Arthritis Program, University Health Network, Toronto, Canada;; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Joan C Fitzgerald
- Regenerative Medicine Institute (REMEDI), National University of Ireland, Galway, Ireland
| | - Frank Barry
- The Arthritis Program, University Health Network, Toronto, Canada;; Regenerative Medicine Institute (REMEDI), National University of Ireland, Galway, Ireland
| | - Sowmya Viswanathan
- The Arthritis Program, University Health Network, Toronto, Canada;; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada; Cell Therapy Program, University Health Network, Toronto, Canada; Division of Hematology, Department of Medicine, University of Toronto, Toronto, Canada.
| |
Collapse
|
658
|
Li X, An G, Wang Y, Liang D, Zhu Z, Tian L. Targeted migration of bone marrow mesenchymal stem cells inhibits silica-induced pulmonary fibrosis in rats. Stem Cell Res Ther 2018; 9:335. [PMID: 30514375 PMCID: PMC6280342 DOI: 10.1186/s13287-018-1083-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/24/2018] [Accepted: 11/19/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Silicosis is a common occupational disease, characterized by silicotic nodules and diffuse pulmonary fibrosis. We demonstrated an anti-fibrotic effect of bone marrow mesenchymal stem cells (BMSCs) in silica-induced lung fibrosis. In the present study, we sought to clarify the homing ability of BMSCs and the specific mechanisms for their effects. METHODS AND RESULTS The biodistribution of BMSCs was identified by near-infrared fluorescence (NIRF) imaging in vivo and in vitro. The results showed that BMSCs labeled with NIR-DiR dyes targeted silica-injured lung tissue, wherein they reached a peak at 6 h post-injection and declined dramatically by day 3. Based on these findings, a second injection of BMSCs was administered 3 days after the first injection. The injected BMSCs migrated to the injured lungs, but did not undergo transformation into specific lung cell types. Interestingly, the injection of BMSC-conditioned medium (BMSCs-CM) significantly attenuated silica-induced pulmonary fibrosis. The collagen deposition and number of nodules were decreased in lung tissues of BMSCs-CM-treated rats. In parallel with these findings, the mRNA levels of collagen I, collagen III, and fibronectin, and the content of transforming growth factor (TGF)-β1 and hydroxyproline were decreased in the BMSCs-CM-treated group compared with the silica group. In addition, alveolar epithelial markers were upregulated by BMSCs-CM treatment. CONCLUSIONS BMSCs migrated to injured areas of the lung after silica instillation and attenuated pulmonary fibrosis. The anti-fibrotic effects of BMSCs were mainly exerted in paracrine manner, rather than through their ability to undergo differentiation.
Collapse
Affiliation(s)
- Xiaoli Li
- Beijing Tropical Medicine Research Institute, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, No. 10, Xi toutiao outside You anmen, Beijing, 100069, China
| | - Guoliang An
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, No. 10, Xi toutiao outside You anmen, Beijing, 100069, China
| | - Yan Wang
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, No. 10, Xi toutiao outside You anmen, Beijing, 100069, China
| | - Di Liang
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, No. 10, Xi toutiao outside You anmen, Beijing, 100069, China
| | - Zhonghui Zhu
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, No. 10, Xi toutiao outside You anmen, Beijing, 100069, China
| | - Lin Tian
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, No. 10, Xi toutiao outside You anmen, Beijing, 100069, China.
| |
Collapse
|
659
|
Ceccato A, Ferrer M, Barbeta E, Torres A. Adjunctive Therapies for Community-Acquired Pneumonia. Clin Chest Med 2018; 39:753-764. [DOI: 10.1016/j.ccm.2018.07.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
660
|
Mehdipour F, Razmkhah M, Rezaeifard S, Bagheri M, Talei A, Khalatbari B, Ghaderi A. Mesenchymal stem cells induced anti‐inflammatory features in B cells from breast tumor draining lymph nodes. Cell Biol Int 2018; 42:1658-1669. [DOI: 10.1002/cbin.11062] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 09/30/2018] [Indexed: 08/30/2023]
Abstract
AbstractThe immune‐modulatory effect of adipose‐derived stem cells (ASCs) on B cells in cancer has not been well elucidated. Herein, the interaction between B cells and ASCs isolated from the breast fat of either normal (nASCs) or breast cancer women (cASCs) was investigated. B cells derived from breast tumor draining lymph nodes were co‐cultured with nASCs or cASCs and B cells proliferation was assessed in direct and transwell assays. Moreover, B cells were co‐cultured with cASCs, nASCs or mesenchymal stromal cells of the tumor tissue (TSCs) and B cell cytokine production was assessed using flow cytometery. cASCs or TSCs were co‐cultured with either intact or B cell depleted lymphocytes and frequencies of CD25+FoxP3+ Tregs, IL‐10+ or IFN‐γ+CD4+ T cells were assessed. Results showed that co‐culture of B cells with ASCs in transwell chambers did not affect B cell proliferation. nASCs, however, was able to significantly reduce B cell proliferation in direct co‐culture experiments (P = 0.004). The frequencies of IL‐10+, TNF‐α+, IL‐2+, and IFN‐γ+ B cells were not significantly different in the co‐cultures of B cells with ASCs or TSCs. But the TNF‐α+/ IL‐10+ B cells ratio decreased in all co‐cultures, a reduction merely significant in B cell‐cASCs co‐culture (P = 0.01). The frequencies of CD4+ T cells subsets in either intact or B cell depleted lymphocytes did not undergo significant changes following co‐culture with ASCs or TSCs. Therefore, ASCs is capable of inhibiting B cell proliferation in a contact dependent manner and shifting the cytokine profile of B cells toward an anti‐inflammatory profile.
Collapse
Affiliation(s)
- Fereshteh Mehdipour
- Shiraz Institute for Cancer Research School of Medicine, Shiraz University of Medical Sciences P.O. Box: 71345‐3119 Shiraz Iran
| | - Mahboobeh Razmkhah
- Shiraz Institute for Cancer Research School of Medicine, Shiraz University of Medical Sciences P.O. Box: 71345‐3119 Shiraz Iran
| | - Somayeh Rezaeifard
- Shiraz Institute for Cancer Research School of Medicine, Shiraz University of Medical Sciences P.O. Box: 71345‐3119 Shiraz Iran
| | | | - Abdol‐Rasoul Talei
- Breast Diseases Research Center Shiraz University of Medical Sciences Shiraz Iran
| | - Behzad Khalatbari
- Department of Plastic Surgery Shiraz University of Medical Sciences Shiraz Iran
| | - Abbas Ghaderi
- Shiraz Institute for Cancer Research School of Medicine, Shiraz University of Medical Sciences P.O. Box: 71345‐3119 Shiraz Iran
- Department of Immunology, School of Medicine Shiraz University of Medical Sciences Shiraz Iran
| |
Collapse
|
661
|
Barreto-Durán E, Mejía-Cruz CC, Leal-García E, Pérez-Núñez R, Rodríguez-Pardo VM. Impact of donor characteristics on the quality of bone marrow as a source of mesenchymal stromal cells. AMERICAN JOURNAL OF STEM CELLS 2018; 7:114-120. [PMID: 30697455 PMCID: PMC6334204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 10/10/2018] [Indexed: 06/09/2023]
Abstract
In recent years, the therapeutic use of mesenchymal stromal cells (MSC) has generated a valuable number of scientific studies that delve into their biological characteristics and their potential in regenerative medicine; however, the impact of the clinical characteristics of tissue donors, from which these cells are isolated, on their potential in applied clinical research is not yet clear. The objective of this study was to evaluate the impact of the clinical characteristics of bone marrow donors on the quality of this tissue as a source of MSC for therapeutic use. Human MSC were isolated, characterized and cultured (according to ISCT criteria) from bone marrow samples from volunteer donors (n = 70) attending the Department of Orthopedics and Traumatology of the Hospital Universitario San Ignacio (Bogota, Colombia) for surgery of prosthetic hip replacement that agreed to participate voluntarily in the study. Donor data such as age, gender, weight, smoker and type of anesthesia used during the surgical procedure were recorded, and the impact of these characteristics on the volume of tissue collection, mononuclear cell count and confluence time of cells with fibroblastoid morphology was evaluated. Correlation coefficients between quantitative variables were calculated with Spearman's correlation test, and the association between qualitative and quantitative variables was evaluated with biserial correlation coefficient. A significant correlation was observed between the age of the donors and the time necessary to obtain confluent cells in vitro (r = 0.2489, P = 0.0377); similarly, the correlation between the volume of bone marrow collected and the number of mononuclear cells obtained was significant (r = 0.7101, P = 0.0001). Although a negative correlation tendency was observed between the mononuclear cell count and the confluence time, this was not significant (r = -0.2041, P = 0.0950). No significant associations were observed between gender, smoking status or type of anesthesia and the expansion characteristics of human mesenchymal stromal cells. Bone marrow donor age and the tissue collection volume impact the time of obtaining MSC in vitro and the mononuclear cell count with which the culture starts. These conditions must be considered when the bone marrow is selected as the tissue for obtaining MSC.
Collapse
Affiliation(s)
- Emilia Barreto-Durán
- Immunobiology and Cell Biology Group, Department of Microbiology, Science Faculty, Pontificia Universidad JaverianaBogotá D.C., Colombia, South America
| | - Claudia Camila Mejía-Cruz
- Immunobiology and Cell Biology Group, Department of Microbiology, Science Faculty, Pontificia Universidad JaverianaBogotá D.C., Colombia, South America
| | - Efrain Leal-García
- Department of Orthopedics and Traumatology, School of Medicine, Pontificia Universidad Javeriana, Hospital Universitario San IgnacioBogotá D.C., Colombia, South America
| | - Rafael Pérez-Núñez
- Department of Orthopedics and Traumatology, School of Medicine, Pontificia Universidad Javeriana, Hospital Universitario San IgnacioBogotá D.C., Colombia, South America
| | - Viviana Marcela Rodríguez-Pardo
- Immunobiology and Cell Biology Group, Department of Microbiology, Science Faculty, Pontificia Universidad JaverianaBogotá D.C., Colombia, South America
| |
Collapse
|
662
|
Salami F, Tavassoli A, Mehrzad J, Parham A. Immunomodulatory effects of mesenchymal stem cells on leukocytes with emphasis on neutrophils. Immunobiology 2018; 223:786-791. [DOI: 10.1016/j.imbio.2018.08.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 08/03/2018] [Indexed: 02/07/2023]
|
663
|
Yousefi F, Lavi Arab F, Saeidi K, Amiri H, Mahmoudi M. Various strategies to improve efficacy of stem cell transplantation in multiple sclerosis: Focus on mesenchymal stem cells and neuroprotection. J Neuroimmunol 2018; 328:20-34. [PMID: 30557687 DOI: 10.1016/j.jneuroim.2018.11.015] [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] [Received: 10/20/2018] [Accepted: 11/30/2018] [Indexed: 02/09/2023]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS) which predominantly affect young adults and undergo heavy socioeconomic burdens. Conventional therapeutic modalities for MS mostly downregulate aggressive immune responses and are almost insufficient for management of progressive course of the disease. Mesenchymal stem cells (MSCs), due to both immunomodulatory and neuroprotective properties have been known as practical cells for treatment of neurodegenerative diseases like MS. However, clinical translation of MSCs is associated with some limitations such as short-life engraftment duration, little in vivo trans-differentiation and restricted accessibility into damaged sites. Therefore, laboratory manipulation of MSCs can improve efficacy of MSCs transplantation in MS patients. In this review, we discuss several novel approaches, which can potentially enhance MSCs capabilities for treating MS.
Collapse
Affiliation(s)
- Forouzan Yousefi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fahimeh Lavi Arab
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Kolsoum Saeidi
- Physiology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Houshang Amiri
- Neurology Research Center, Kerman University of Medical Sciences, Kerman, Iran; Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Mahmoud Mahmoudi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
664
|
Najar M, Fayyad-Kazan H, Faour WH, Merimi M, Sokal EM, Lombard CA, Fahmi H. Immunological modulation following bone marrow-derived mesenchymal stromal cells and Th17 lymphocyte co-cultures. Inflamm Res 2018; 68:203-213. [PMID: 30506263 DOI: 10.1007/s00011-018-1205-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 11/16/2018] [Accepted: 11/23/2018] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE AND DESIGN The objective of the study is to uncover the influence of human bone marrow-derived mesenchymal stem cells (BM-MSCs) on the generation of Th17 lymphocytes in co-cultures of both BM-MSCs and T cells. MATERIALS AND METHODS BM-MSCs, characterized according to the international society for cellular therapy (ISCT) criteria, were co-cultured with T cells isolated from peripheral blood. The expression levels of IL-17 receptor, RORγt and IL-23 receptor were evaluated using flow cytometry. The levels of cytokines involved in Th17 immunomodulation were measured using multiplex assay. TREATMENT Inflammatory primed and non-primed BM-MSCs were co-cultured with either activated or non-activated T cells either at (1/80) and (1/5) ratio respectively. RESULTS MSC/T-cell ratio and inflammation significantly influenced the effect of BM-MSCs on the generation of Th17 lymphocytes. Cocultures of either primed or non-primed BM-MSCs with activated T cells significantly induced IL-17A-expressing lymphocytes. Interestingly, the expression of the transcription factor RORγt was significantly increased when compared to levels in activated T cells. Finally, both cell ratio and priming of BM-MSCs with cytokines substantially influenced the cytokine profile of BM-MSCs and T cells. CONCLUSION Our findings suggest that BM-MSCs significantly modulate the Th17 lymphocyte pathway in a complex manner.
Collapse
Affiliation(s)
- Mehdi Najar
- Osteoarthritis Research Unit, Department of Medicine, University of Montreal Hospital Research Center (CRCHUM), 900 rue Saint-Denis, R11.424, Montreal, QC, H2X 0A9, Canada
| | - Hussein Fayyad-Kazan
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences I, Lebanese University, Hadath, Lebanon
| | - Wissam H Faour
- Pharmacology, Gilbert and Rose-Mary Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon.
| | - Makram Merimi
- Laboratory of Experimental Hematology, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
- Laboratory of Physiology, Ethnopharmacology and Genetics, Faculty of Sciences, University Mohammed Premier, Oujda, Morocco
| | - Etienne M Sokal
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale and Clinique (IREC), Université Catholique de Louvain, 1200, Brussels, Belgium
| | - Catherine A Lombard
- Laboratory of Pediatric Hepatology and Cell Therapy, Institut de Recherche Expérimentale and Clinique (IREC), Université Catholique de Louvain, 1200, Brussels, Belgium
| | - Hassan Fahmi
- Osteoarthritis Research Unit, Department of Medicine, University of Montreal Hospital Research Center (CRCHUM), 900 rue Saint-Denis, R11.424, Montreal, QC, H2X 0A9, Canada
| |
Collapse
|
665
|
Bandeiras C, Cabral JM, Finkelstein SN, Ferreira FC. Modeling biological and economic uncertainty on cell therapy manufacturing: the choice of culture media supplementation. Regen Med 2018; 13:917-933. [PMID: 30488770 DOI: 10.2217/rme-2018-0034] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
AIM To evaluate the cost-effectiveness of autologous cell therapy manufacturing in xeno-free conditions. MATERIALS & METHODS Published data on the isolation and expansion of mesenchymal stem/stromal cells introduced donor, multipassage and culture media variability on cell yields and process times on adherent culture flasks to drive cost simulation of a scale-out campaign of 1000 doses of 75 million cells each in a 400 square meter Good Manufacturing Practices facility. RESULTS & CONCLUSION Passage numbers in the expansion step are strongly associated with isolation cell yield and drive cost increases per donor of $1970 and 2802 for fetal bovine serum and human platelet lysate. Human platelet lysate decreases passage numbers and process costs in 94.5 and 97% of donors through lower facility and labor costs. Cost savings are maintained with full equipment depreciation and higher numbers of cells per dose, highlighting the number of cells per passage step as the key cost driver.
Collapse
Affiliation(s)
- Cátia Bandeiras
- Department of Bioengineering and iBB - Institute for Bioengineering & Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal.,The Discoveries Centre for Regenerative & Precision Medicine, Lisbon Campus, Universidade de Lisboa, Portugal.,Institute for Data, Systems & Society, Massachusetts Institute of Technology, 50 Ames Street, Cambridge MA 02139, USA.,Division of Clinical Informatics, Department of Medicine, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston MA 02215, USA
| | - Joaquim Ms Cabral
- Department of Bioengineering and iBB - Institute for Bioengineering & Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal.,The Discoveries Centre for Regenerative & Precision Medicine, Lisbon Campus, Universidade de Lisboa, Portugal
| | - Stan N Finkelstein
- Institute for Data, Systems & Society, Massachusetts Institute of Technology, 50 Ames Street, Cambridge MA 02139, USA.,Division of Clinical Informatics, Department of Medicine, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston MA 02215, USA
| | - Frederico Castelo Ferreira
- Department of Bioengineering and iBB - Institute for Bioengineering & Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal.,The Discoveries Centre for Regenerative & Precision Medicine, Lisbon Campus, Universidade de Lisboa, Portugal
| |
Collapse
|
666
|
Learn GD, McClellan PE, Knapik DM, Cumsky JL, Webster-Wood V, Anderson JM, Gillespie RJ, Akkus O. Woven collagen biotextiles enable mechanically functional rotator cuff tendon regeneration during repair of segmental tendon defects in vivo. J Biomed Mater Res B Appl Biomater 2018; 107:1864-1876. [PMID: 30485649 DOI: 10.1002/jbm.b.34279] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 10/04/2018] [Accepted: 10/19/2018] [Indexed: 12/15/2022]
Abstract
Despite advancements in surgical techniques and materials for rotator cuff repair procedures, primary repair failures remain common. This study examines the use of electrochemically aligned collagen (ELAC) threads woven into biotextile scaffolds as grafts to repair critical infraspinatus tendon defects in New Zealand White rabbits. Three surgical treatment groups were evaluated: rabbits undergoing direct repair as operative controls, rabbits receiving ELAC scaffolds alone, and rabbits treated with mesenchymal stem cell (MSC)-seeded ELAC scaffolds. In each animal, the intact, contralateral infraspinatus served as an internal positive control. Tendon-bone constructs were harvested after 3 months in vivo and outcome measures included biomechanical testing, histological staining, and immunohistochemical staining. Biomechanical testing revealed that maximum load-bearing capacity was comparable between all groups, while MSC-seeded scaffold repairs exhibited increased stiffness relative to non-seeded scaffold repairs. Histological staining revealed robust collagen deposition around ELAC fibers and increased cellularity within the continuum of woven scaffolds as compared to native tendon. Immunohistochemical staining revealed presence of collagens I and III in all groups, but procollagen I and the tendon-specific marker tenomodulin were only observed in seeded and non-seeded ELAC scaffold repairs. Findings of this pilot study warrant continued investigation of ELAC biotextile scaffolds for repair of critically-sized rotator cuff tendon defects. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B: 1864-1876, 2019.
Collapse
Affiliation(s)
- Greg D Learn
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Phillip E McClellan
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Derrick M Knapik
- Department of Orthopaedic Surgery, University Hospitals of Cleveland, Cleveland, Ohio
| | - Jameson L Cumsky
- School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Victoria Webster-Wood
- Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, Ohio
| | - James M Anderson
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio.,Department of Pathology, Case Western Reserve University, Cleveland, Ohio.,Department of Macromolecular Science, Case Western Reserve University, Cleveland, Ohio
| | - Robert J Gillespie
- Department of Orthopaedic Surgery, University Hospitals of Cleveland, Cleveland, Ohio
| | - Ozan Akkus
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio.,Department of Mechanical and Aerospace Engineering, Case Western Reserve University, Cleveland, Ohio.,Department of Orthopaedic Surgery, University Hospitals of Cleveland, Cleveland, Ohio
| |
Collapse
|
667
|
Scarfe L, Taylor A, Sharkey J, Harwood R, Barrow M, Comenge J, Beeken L, Astley C, Santeramo I, Hutchinson C, Ressel L, Smythe J, Austin E, Levy R, Rosseinsky MJ, Adams DJ, Poptani H, Park BK, Murray P, Wilm B. Non-invasive imaging reveals conditions that impact distribution and persistence of cells after in vivo administration. Stem Cell Res Ther 2018; 9:332. [PMID: 30486897 PMCID: PMC6264053 DOI: 10.1186/s13287-018-1076-x] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 10/23/2018] [Accepted: 11/12/2018] [Indexed: 12/13/2022] Open
Abstract
Background Cell-based regenerative medicine therapies are now frequently tested in clinical trials. In many conditions, cell therapies are administered systemically, but there is little understanding of their fate, and adverse events are often under-reported. Currently, it is only possible to assess safety and fate of cell therapies in preclinical studies, specifically by monitoring animals longitudinally using multi-modal imaging approaches. Here, using a suite of in vivo imaging modalities to explore the fate of a range of human and murine cells, we investigate how route of administration, cell type and host immune status affect the fate of administered cells. Methods We applied a unique imaging platform combining bioluminescence, optoacoustic and magnetic resonance imaging modalities to assess the safety of different human and murine cell types by following their biodistribution and persistence in mice following administration into the venous or arterial system. Results Longitudinal imaging analyses (i) suggested that the intra-arterial route may be more hazardous than intravenous administration for certain cell types, (ii) revealed that the potential of a mouse mesenchymal stem/stromal cell (MSC) line to form tumours depended on administration route and mouse strain and (iii) indicated that clinically tested human umbilical cord (hUC)-derived MSCs can transiently and unexpectedly proliferate when administered intravenously to mice. Conclusions In order to perform an adequate safety assessment of potential cell-based therapies, a thorough understanding of cell biodistribution and fate post administration is required. The non-invasive imaging platform used here can expose not only the general organ distribution of these therapies, but also a detailed view of their presence within different organs and, importantly, tumourigenic potential. Our observation that the hUC-MSCs but not the human bone marrow (hBM)-derived MSCs persisted for a period in some animals suggests that therapies with these cells should proceed with caution. Electronic supplementary material The online version of this article (10.1186/s13287-018-1076-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lauren Scarfe
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK.,Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Arthur Taylor
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK.,Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Jack Sharkey
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK.,Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Rachel Harwood
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK.,Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Michael Barrow
- Department of Chemistry, University of Liverpool, Liverpool, UK
| | - Joan Comenge
- Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Lydia Beeken
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK
| | - Cai Astley
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK
| | - Ilaria Santeramo
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK.,Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Claire Hutchinson
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Lorenzo Ressel
- Department of Veterinary Pathology and Public Health, Institute of Veterinary Science, University of Liverpool, Liverpool, UK
| | | | | | - Raphael Levy
- Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | | | - Dave J Adams
- School of Chemistry, College of Science and Engineering, University of Glasgow, Glasgow, UK
| | - Harish Poptani
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK.,Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK
| | - Brian K Park
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Patricia Murray
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK. .,Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK. .,Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK.
| | - Bettina Wilm
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, UK. .,Centre for Preclinical Imaging, University of Liverpool, Liverpool, UK. .,Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK.
| |
Collapse
|
668
|
Enhancement of the efficacy of mesenchymal stem cells in the treatment of ischemic diseases. Biomed Pharmacother 2018; 109:2022-2034. [PMID: 30551458 DOI: 10.1016/j.biopha.2018.11.068] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 11/17/2018] [Accepted: 11/19/2018] [Indexed: 02/05/2023] Open
Abstract
Ischemic diseases refer to a wide range of diseases caused by reduced blood flow and a subsequently deficient oxygen and nutrient supply. The pathogenesis of ischemia is multifaceted and primarily involves inflammation, oxidative stress and an apoptotic response. Over the last decade, mesenchymal stem cells (MSCs) have been widely studied as potential cell therapy agents for ischemic diseases due to their multiple favourable functions. However, the low homing and survival rates of transplanted cells have been concerns limiting for their clinical application. Recently, increasing studies have attempted to enhance the efficacy of MSCs by various strategies including genetic modification, pretreatment, combined application and biomaterial application. The purpose of this review is to summarize these creative strategies and the progress in basic and preclinical studies.
Collapse
|
669
|
Sheykhhasan M, Manoochehri H, Pourjafar M, Fayazi N. Mesenchymal stem cells as a valuable agent in osteoarthritis treatment. Stem Cell Investig 2018; 5:41. [PMID: 30596081 PMCID: PMC6286892 DOI: 10.21037/sci.2018.11.04] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Accepted: 11/11/2018] [Indexed: 02/05/2023]
Affiliation(s)
- Mohsen Sheykhhasan
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Mesenchymal Stem Cell, the Academic Center for Education, Culture and Research, Qom Branch, Qom, Iran
| | - Hamed Manoochehri
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mona Pourjafar
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Nashmin Fayazi
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
670
|
An JH, Song WJ, Li Q, Kim SM, Yang JI, Ryu MO, Nam AR, Bhang DH, Jung YC, Youn HY. Prostaglandin E 2 secreted from feline adipose tissue-derived mesenchymal stem cells alleviate DSS-induced colitis by increasing regulatory T cells in mice. BMC Vet Res 2018; 14:354. [PMID: 30453939 PMCID: PMC6245895 DOI: 10.1186/s12917-018-1684-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 11/01/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is an intractable autoimmune disease, relatively common in cats, with chronic vomiting and diarrhea. Previous studies have reported that mesenchymal stem cells (MSCs) alleviate inflammation by modulating immune cells. However, there is a lack of research on cross-talk mechanism between feline adipose tissue-derived mesenchymal stem cells (fAT-MSCs) and immune cells in IBD model. Hence, this study aimed to evaluate the therapeutic effects of fAT-MSC on mice model of colitis and to clarify the therapeutic mechanism of fAT-MSCs. RESULTS Intraperitoneal infusion of fAT-MSC ameliorated the clinical and histopathologic severity of colitis, including body weight loss, diarrhea, and inflammation in the colon of Dextran sulfate sodium (DSS)-treated mice (C57BL/6). Since regulatory T cells (Tregs) are pivotal in modulating immune responses and maintaining tolerance in colitis, the relation of Tregs with fAT-MSC-secreted factor was investigated in vitro. PGE2 secreted from fAT-MSC was demonstrated to induce elevation of FOXP3 mRNA expression and adjust inflammatory cytokines in Con A-induced feline peripheral blood mononuclear cells (PBMCs). Furthermore, in vivo, FOXP3+ cells of the fAT-MSC group were significantly increased in the inflamed colon, relative to that in the PBS group. CONCLUSION Our results suggest that PGE2 secreted from fAT-MSC can reduce inflammation by increasing FOXP3+ Tregs in mice model of colitis. Consequently, these results propose the possibility of administration of fAT-MSC to cats with not only IBD but also other immune-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Ju-Hyun An
- Labolatory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Woo-Jin Song
- Labolatory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Qiang Li
- Labolatory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Sang-Min Kim
- Labolatory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Ji-In Yang
- Labolatory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Min-Ok Ryu
- Labolatory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - A Ryung Nam
- Labolatory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Dong Ha Bhang
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon-si, Gyeonggi-do, 16419, Republic of Korea
| | - Yun-Chan Jung
- Chaon Corporation, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13493, Republic of Korea
| | - Hwa-Young Youn
- Labolatory of Veterinary Internal Medicine, Department of Veterinary Clinical Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.
| |
Collapse
|
671
|
Wang K, Li Z, Li J, Liao W, Qin Y, Zhang N, Huo X, Mao N, Zhu H. Optimization of the Platelet-Rich Plasma Concentration for Mesenchymal Stem Cell Applications. Tissue Eng Part A 2018; 25:333-351. [PMID: 30234444 DOI: 10.1089/ten.tea.2018.0091] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
IMPACT STATEMENT In the current study, we screened the effects of different concentrations of platelet-rich plasma (PRP) on the mesenchymal stem cell (MSC) proliferation and differentiation. Our data demonstrated that concentrated PRP exerted different effects on cell proliferation and on the osteogenic, adipogenic, and chondrogenic differentiation of human bone marrow-derived MSCs. Meanwhile, we optimized PRP concentrations for each of these lineage differentiation and MSC expansion and, thus, suggested several promising clinical strategies for different kinds of diseases. These findings may help explain the controversial effects of PRP on MSCs and improve the progress in precise applications of PRP-based regenerative strategies.
Collapse
Affiliation(s)
- Ketao Wang
- 1 Department of Orthopedics, Center of Sport Medicine, Chinese PLA General Hospital, Beijing, China
| | - Zhongli Li
- 1 Department of Orthopedics, Center of Sport Medicine, Chinese PLA General Hospital, Beijing, China
| | - Ji Li
- 1 Department of Orthopedics, Center of Sport Medicine, Chinese PLA General Hospital, Beijing, China
| | - Weixiong Liao
- 1 Department of Orthopedics, Center of Sport Medicine, Chinese PLA General Hospital, Beijing, China
| | - Yuanyuan Qin
- 2 Department of Blood Transfusion, Chinese PLA General Hospital, Beijing, China
| | - Ning Zhang
- 1 Department of Orthopedics, Center of Sport Medicine, Chinese PLA General Hospital, Beijing, China
| | - Xiulin Huo
- 1 Department of Orthopedics, Center of Sport Medicine, Chinese PLA General Hospital, Beijing, China
| | - Ning Mao
- 3 Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Heng Zhu
- 3 Beijing Institute of Basic Medical Sciences, Beijing, China
| |
Collapse
|
672
|
Rezaei F, Moazzeni SM. Comparison of The Therapeutic Effect of Syngeneic, Allogeneic, and Xenogeneic Adipose Tissue-Derived Mesenchymal Stem Cells on Abortion Rates in A Mouse Model. CELL JOURNAL 2018; 21:92-98. [PMID: 30507094 PMCID: PMC6275426 DOI: 10.22074/cellj.2019.5954] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 06/03/2018] [Indexed: 12/15/2022]
Abstract
Objective Mesenchymal stem cells (MSCs), due to their immunomodulatory functions, are an ideal candidate
for the treatment of immune-related diseases. Recurrent spontaneous abortion (RSA) is one of the most common
complications of pregnancy which in many cases is related to the immune system disorders. Our previous study has
shown that the abortion rate was decreased following the syngeneic MSCs therapy in abortion-prone mice. In this
study, the therapeutic effect of syngeneic, allogeneic, and xenogeneic MSCs was compared in a mouse model of RSA.
Materials and Methods In this experimental study, MSCs were isolated from adipose tissue (ASCs) of CBA/J and
BALB/c mice and human. After characterization, ASCs were injected (IP) at day 4 of gestation to female CBA/J mice
following their mating with DBA/2 male mice. In the control group, phosphate-buffered saline (PBS) was injected and
CBA/J×BALB/c mating was also used as the normal pregnancy control. On day 14.5 of pregnancy, embryo resorption
rate was determined.
Results The abortion rate significantly decreased following the ASCs therapy from syngeneic (6.31%), allogeneic
(6.54%), and xenogeneic group (12.36%) compared to ASCs non-treated group (34.4%). There was no statistical
difference between ASCs treated groups, however syngeneic and allogeneic ASCs reduced the abortion rate more
efficiently than xenogeneic ASC.
Conclusion The abortion rate was significantly decreased following the intraperitoneal administration of ASCs from
various donated sources in abortion-prone mice. These results indicated that the immunogenicity of allogeneic and
xenogeneic ASCs is not a contradictory problem for their therapeutic effects on RSA.
Collapse
Affiliation(s)
- Fatemeh Rezaei
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Seyed Mohammad Moazzeni
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran. Electronic Address:
| |
Collapse
|
673
|
Mardpour S, Hamidieh AA, Taleahmad S, Sharifzad F, Taghikhani A, Baharvand H. Interaction between mesenchymal stromal cell-derived extracellular vesicles and immune cells by distinct protein content. J Cell Physiol 2018; 234:8249-8258. [PMID: 30378105 DOI: 10.1002/jcp.27669] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 10/02/2018] [Indexed: 12/13/2022]
Abstract
Mesenchymal stromal cells (MSCs) can effectively contribute to tissue regeneration inside the inflammatory microenvironment mostly through modulating immune responses. MSC-derived extracellular vesicles (MSC-EVs) display immunoregulatory functions similar to parent cells. Interactions between MSC-EVs and immune cells make them an ideal therapeutic candidate for infectious, inflammatory, and autoimmune diseases. These properties of MSC-EVs have encouraged researchers to perform extensive studies on multiple factors that mediate MSC-EVs immunomodulatory effects. Investigation of proteins involved in the complex interplay of MSC-EVs and immune cells may help us to better understand their functions. Here, we performed a comprehensive proteomic analysis of MSC-EVs that was previously reported by ExoCarta database. A total of 938 proteins were identified as MSC-EV proteome using quantitative proteomics techniques. Kyoto Encyclopedia of Genes and Genomes analysis demonstrates that ECM-receptor interaction, focal adhesion, and disease-specific pathways are enriched in MSC-EVs. By detail analysis of proteins presence in immune system process, we found that expression of some cytokines, chemokines, and chemokine receptors such as IL10, HGF, LIF, CCL2, VEGFC, and CCL20, which leads to migration of MSC-EVs to injured sites, suppression of inflammation and promotion of regeneration in inflammatory and autoimmune diseases. Also, some chemoattractant proteins such as CXCL2, CXCL8, CXCL16, DEFA1, HERC5, and IFITM2 were found in MSC-EV proteome. They may actively recruit immune cells to the proximity of MSC or MSC-EVs, may result in boosting immune response under specific circumstances, and may have protective role in infectious diseases. In this review, we summarize available information about immunomodulation of MSC-EVs with particular emphasis on their proteomics analysis.
Collapse
Affiliation(s)
- Soura Mardpour
- Tissue Engineering and Applied Cell Sciences Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Amir Ali Hamidieh
- Pediatric Stem Cell Transplant Department, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Sara Taleahmad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Farzaneh Sharifzad
- Department of Applied Cell Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Adeleh Taghikhani
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modarres University, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| |
Collapse
|
674
|
D’Angelo W, Chen B, Gurung C, Guo YL. Characterization of embryonic stem cell-differentiated fibroblasts as mesenchymal stem cells with robust expansion capacity and attenuated innate immunity. Stem Cell Res Ther 2018; 9:278. [PMID: 30359317 PMCID: PMC6203291 DOI: 10.1186/s13287-018-1033-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/14/2018] [Accepted: 09/30/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) isolated from adult tissues (Ad-MSCs) have shown great promise for use in regenerative medicine. However, their poor in vitro expansion capacity and tissue scarcity have been major limitations. In this study, we demonstrate that mouse embryonic stem cells (mESCs) can differentiate into cells with MSC properties. METHODS Using previously established methods that characterize Ad-MSCs, we analyzed mESC-differentiated fibroblasts (mESC-FBs), including plastic adherence, clonogenic growth, MSC marker expression, tri-lineage differentiation potential, and the capacity to express immunomodulators. RESULTS Although previously characterized as mESC-differentiated fibroblasts (mESC-FBs), these cells exhibit major properties of Ad-MSCs. However, mESC-FBs also display unique features inherited from ESCs, including robust expansion capacity, senescence resistance, and attenuated innate immunity. In particular, mESC-FBs are insensitive to bacterial endotoxin (lipopolysaccharide, LPS) and do not express LPS-induced inflammatory molecules, in contrast to bone marrow (BM)-MSCs. We further demonstrate that mESC-FBs are resistant to the cytotoxicity associated with inflammatory cytokines, bacterial endotoxins (LPS and heat-killed bacteria), and macrophage-mediated inflammation. CONCLUSIONS While it remains to be determined how the unique properties of mESC-FBs will affect their immunoregulatory activity under an in vivo condition, our findings demonstrate that ESCs could be used as an alternative source to generate a new class of ESC-MSCs with unique features potentially useful in regenerative medicine.
Collapse
Affiliation(s)
- William D’Angelo
- Department of Cell and Molecular Biology, University of Southern Mississippi, 118 College Drive 5018, Hattiesburg, MS 39406 USA
| | - Bohan Chen
- Department of Cell and Molecular Biology, University of Southern Mississippi, 118 College Drive 5018, Hattiesburg, MS 39406 USA
| | - Chandan Gurung
- Department of Cell and Molecular Biology, University of Southern Mississippi, 118 College Drive 5018, Hattiesburg, MS 39406 USA
| | - Yan-Lin Guo
- Department of Cell and Molecular Biology, University of Southern Mississippi, 118 College Drive 5018, Hattiesburg, MS 39406 USA
| |
Collapse
|
675
|
Zhang Y, Xing Y, Jia L, Ji Y, Zhao B, Wen Y, Xu X. An In Vitro Comparative Study of Multisource Derived Human Mesenchymal Stem Cells for Bone Tissue Engineering. Stem Cells Dev 2018; 27:1634-1645. [PMID: 30234437 DOI: 10.1089/scd.2018.0119] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have been considered promising tools for tissue engineering and regenerative medicine. However, the optimal cell source for bone regeneration remains controversial. To better identify seed cells for bone tissue engineering, we compared MSCs from seven different tissues, including four from dental origins, dental pulp stem cells (DPSCs), periodontal ligament stem cells (PDLSCs), gingival MSCs (GMSCs), and dental follicle stem cells (DFSCs); two from somatic origins, bone marrow-derived MSCs (BM-MSCs) and adipose-derived stem cells (ADSCs); and one from birth-associated perinatal tissue umbilical cord (UCMSCs). We cultured the cells under a standardized culture condition and studied their biological characteristics. According to our results, these cells exhibited similar immunophenotype and had potential for multilineage differentiation. MSCs from dental and perinatal tissues proliferated more rapidly than those from somatic origins. Simultaneously, DPSCs and PDLSCs owned stronger antiapoptotic ability under the microenvironment of oxidative stress combined with serum deprivation. In respect to osteogenic differentiation, the two somatic MSCs, BM-MSCs and ADSCs, demonstrated the strongest ability for osteogenesis compared to PDLSCs and DFSCs, which were just a little bit weaker than the formers. However, GMSCs and UCMSCs were the most pertinacious ones to differentiate to osteoblasts. We also revealed that the canonical intracellular protein kinase-based cascade signaling pathways, including PI3K/AKT, MAPK/ERK, and p38 MAPK, possessed different levels of activation in different MSCs after osteoblast induction. Our conclusions suggest that PDLSCs might be a good potential alternative to BM-MSCs for bone tissue engineering.
Collapse
Affiliation(s)
- Yunpeng Zhang
- 1 School of Stomatology, Shandong University , Jinan, P.R. China .,2 Shandong Provincial Key Laboratory of Oral Tissue Regeneration , Jinan, P.R. China
| | - Yixiao Xing
- 1 School of Stomatology, Shandong University , Jinan, P.R. China .,2 Shandong Provincial Key Laboratory of Oral Tissue Regeneration , Jinan, P.R. China
| | - Linglu Jia
- 1 School of Stomatology, Shandong University , Jinan, P.R. China .,2 Shandong Provincial Key Laboratory of Oral Tissue Regeneration , Jinan, P.R. China
| | - Yawen Ji
- 1 School of Stomatology, Shandong University , Jinan, P.R. China .,2 Shandong Provincial Key Laboratory of Oral Tissue Regeneration , Jinan, P.R. China
| | - Bin Zhao
- 1 School of Stomatology, Shandong University , Jinan, P.R. China .,2 Shandong Provincial Key Laboratory of Oral Tissue Regeneration , Jinan, P.R. China
| | - Yong Wen
- 1 School of Stomatology, Shandong University , Jinan, P.R. China .,2 Shandong Provincial Key Laboratory of Oral Tissue Regeneration , Jinan, P.R. China
| | - Xin Xu
- 1 School of Stomatology, Shandong University , Jinan, P.R. China .,2 Shandong Provincial Key Laboratory of Oral Tissue Regeneration , Jinan, P.R. China
| |
Collapse
|
676
|
Kusamori K, Takayama Y, Nishikawa M. Stable Surface Modification of Mesenchymal Stem Cells Using the Avidin-Biotin Complex Technique. ACTA ACUST UNITED AC 2018; 47:e66. [DOI: 10.1002/cpsc.66] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Kosuke Kusamori
- Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science; Chiba Japan
| | - Yukiya Takayama
- Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science; Chiba Japan
| | - Makiya Nishikawa
- Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science; Chiba Japan
| |
Collapse
|
677
|
Nugraha AP, Narmada IB, Ernawati DS, Dinaryanti A, Hendrianto E, Riawan W, Rantam FA. Bone alkaline phosphatase and osteocalcin expression of rat's Gingival mesenchymal stem cells cultured in platelet-rich fibrin for bone remodeling ( in vitro study). Eur J Dent 2018; 12:566-573. [PMID: 30369804 PMCID: PMC6178667 DOI: 10.4103/ejd.ejd_261_18] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE The aim of this study was to analyze the osteogenic differentiation of rat GMSCs cultured in PRF for bone remodeling. MATERIALS AND METHODS GMSCs were isolated from the lower gingival tissue of four healthy, 250 g, 1-month old, male rats (Rattus norvegicus) cut into small fragments, cultured for 2 weeks, and subsequently passaged every 4-5 days. GMSCs isolated in passage 3 were characterized by CD34, CD45, CD44, CD73, CD90, and CD105 using fluorescein isothiocyanate immunocytochemistry (ICC) examination. GMSCs in passage 3-5 cultured in five M24 plates (N = 108; n = 6/group) for 7, 14, and 21 days with three different mediums as follows: Control (-) group: α-Modified Eagle Medium; Control (+) group: High-dose glucose Dulbecco's Modified Eagle's Medium (DMEM-HG) + osteogenic medium; and treatment group: DMEM-HG + osteogenic medium + PRF. GMSCs were osteogenic differentiation cultured in vitro in three different mediums by bone alkaline phosphatase (BALP) and osteocalcin (OSC) marker using ICC monoclonal antibody. STATISTICAL ANALYSIS USED The one-way analysis of variance was performed (P < 0.05) based on Shapiro-Wilk and Levene's tests (P > 0.05). RESULTS GMSCs were shown to present + CD44, +CD73, +CD90, +CD105 and - CD34, - and CD45 expression as MSCs markers. The treatment group showed the highest BALP expression (16.00 ± 1.732) on day 7, while OSC expression (13.67 ± 2.309) on day 21 showed the statistically significant difference between groups (P < 0.05). CONCLUSION GMSCs cultured in PRF demonstrated potential osteogenic differentiation ability capable of accelerating in vitro bone remodeling by enhancing BALP and OSC expression.
Collapse
Affiliation(s)
- Alexander Patera Nugraha
- Department of Orthodontic, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
- Doctoral Student of Medical Science, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Stem Cell Research and Development Center, Universitas Airlangga, Surabaya, Indonesia
| | - Ida Bagus Narmada
- Department of Orthodontic, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Diah Savitri Ernawati
- Department of Oral Medicine Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Aristika Dinaryanti
- Stem Cell Research and Development Center, Universitas Airlangga, Surabaya, Indonesia
| | - Eryk Hendrianto
- Stem Cell Research and Development Center, Universitas Airlangga, Surabaya, Indonesia
| | - Wibi Riawan
- Department of Biochemistry and Molecular, Biochemistry Biomolecular Laboratory, Faculty of Medicine, Universitas Brawijaya, Surabaya, Indonesia
| | - Fedik Abdul Rantam
- Stem Cell Research and Development Center, Universitas Airlangga, Surabaya, Indonesia
- Department of Microbiology, Virology and Immunology Laboratory, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| |
Collapse
|
678
|
Bahsoun S, Coopman K, Forsyth NR, Akam EC. The Role of Dissolved Oxygen Levels on Human Mesenchymal Stem Cell Culture Success, Regulatory Compliance, and Therapeutic Potential. Stem Cells Dev 2018; 27:1303-1321. [DOI: 10.1089/scd.2017.0291] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Soukaina Bahsoun
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, United Kingdom
| | - Karen Coopman
- Centre for Biological Engineering, Loughborough University, Loughborough, United Kingdom
| | - Nicholas R. Forsyth
- Guy Hilton Research Centre, Institute for Science and Technology in Medicine, Keele University, Keele, United Kingdom
| | - Elizabeth C. Akam
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, United Kingdom
| |
Collapse
|
679
|
Yao J, Zheng J, Cai J, Zeng K, Zhou C, Zhang J, Li S, Li H, Chen L, He L, Chen H, Fu H, Zhang Q, Chen G, Yang Y, Zhang Y. Extracellular vesicles derived from human umbilical cord mesenchymal stem cells alleviate rat hepatic ischemia-reperfusion injury by suppressing oxidative stress and neutrophil inflammatory response. FASEB J 2018; 33:1695-1710. [PMID: 30226809 DOI: 10.1096/fj.201800131rr] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cells (MSCs) have been reported to exert therapeutic effects on immunoregulation, tissue repair, and regeneration from the bench to the bedside. Increasing evidence demonstrates that extracellular vesicles (EVs) derived from MSCs could contribute to these effects and are considered as a potential replacement for stem cell-based therapies. However, the efficacy and underlying mechanisms of EV-based treatment in hepatic ischemia-reperfusion injury (IRI) remain unclear. Here, we demonstrated that human umbilical cord MSC-EVs (huc-MSC-EVs) could protect against IRI-induced hepatic apoptosis by reducing the infiltration of neutrophils and alleviating oxidative stress in hepatic tissue in vivo. Meanwhile, huc-MSC-EVs reduced the respiratory burst of neutrophils and prevented hepatocytes from oxidative stress-induced cell death in vitro. Interestingly, we found that the mitochondria-located antioxidant enzyme, manganese superoxide dismutase (MnSOD), was encapsulated in huc-MSC-EVs and reduced oxidative stress in the hepatic IRI model. Knockdown of MnSOD in huc-MSCs decreased the level of MnSOD in huc-MSC-EVs and attenuated the antiapoptotic and antioxidant capacities of huc-MSC-EVs, which could be partially rescued by MnSOD mimetic manganese (III) 5,10,15,20-tetrakis (4-benzoic acid) porphyrin (MnTBAP). In summary, these findings provide new clues to reveal the therapeutic effects of huc-MSC-EVs on hepatic IRI and evaluate their preclinical application.-Yao, J., Zheng, J., Cai, J., Zeng, K., Zhou, C., Zhang, J., Li, S., Li, H., Chen, L., He, L., Chen, H., Fu, H., Zhang, Q., Chen, G., Yang, Y., Zhang, Y. Extracellular vesicles derived from human umbilical cord mesenchymal stem cells alleviate rat hepatic ischemia-reperfusion injury by suppressing oxidative stress and neutrophil inflammatory response.
Collapse
Affiliation(s)
- Jia Yao
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital, Organ Transplantation Research Center of Guangdong Province, Organ Transplantation Institute, Sun Yat-sen University, Guangzhou, China.,Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine, Guangdong Higher Education Institutes, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jun Zheng
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital, Organ Transplantation Research Center of Guangdong Province, Organ Transplantation Institute, Sun Yat-sen University, Guangzhou, China.,Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine, Guangdong Higher Education Institutes, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianye Cai
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital, Organ Transplantation Research Center of Guangdong Province, Organ Transplantation Institute, Sun Yat-sen University, Guangzhou, China.,Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine, Guangdong Higher Education Institutes, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kaining Zeng
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital, Organ Transplantation Research Center of Guangdong Province, Organ Transplantation Institute, Sun Yat-sen University, Guangzhou, China.,Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine, Guangdong Higher Education Institutes, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chaorong Zhou
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital, Organ Transplantation Research Center of Guangdong Province, Organ Transplantation Institute, Sun Yat-sen University, Guangzhou, China.,Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine, Guangdong Higher Education Institutes, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiebin Zhang
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital, Organ Transplantation Research Center of Guangdong Province, Organ Transplantation Institute, Sun Yat-sen University, Guangzhou, China.,Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine, Guangdong Higher Education Institutes, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shihui Li
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital, Organ Transplantation Research Center of Guangdong Province, Organ Transplantation Institute, Sun Yat-sen University, Guangzhou, China
| | - Hui Li
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital, Organ Transplantation Research Center of Guangdong Province, Organ Transplantation Institute, Sun Yat-sen University, Guangzhou, China.,Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine, Guangdong Higher Education Institutes, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liang Chen
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital, Organ Transplantation Research Center of Guangdong Province, Organ Transplantation Institute, Sun Yat-sen University, Guangzhou, China.,Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine, Guangdong Higher Education Institutes, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liying He
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Huaxin Chen
- Cell-Gene Therapy Translational Medicine Research Center, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hongyuan Fu
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital, Organ Transplantation Research Center of Guangdong Province, Organ Transplantation Institute, Sun Yat-sen University, Guangzhou, China.,Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine, Guangdong Higher Education Institutes, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qi Zhang
- Cell-Gene Therapy Translational Medicine Research Center, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guihua Chen
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital, Organ Transplantation Research Center of Guangdong Province, Organ Transplantation Institute, Sun Yat-sen University, Guangzhou, China.,Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine, Guangdong Higher Education Institutes, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yang Yang
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital, Organ Transplantation Research Center of Guangdong Province, Organ Transplantation Institute, Sun Yat-sen University, Guangzhou, China.,Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine, Guangdong Higher Education Institutes, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yingcai Zhang
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital, Organ Transplantation Research Center of Guangdong Province, Organ Transplantation Institute, Sun Yat-sen University, Guangzhou, China.,Guangdong Key Laboratory of Liver Disease Research, Key Laboratory of Liver Disease Biotherapy and Translational Medicine, Guangdong Higher Education Institutes, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
680
|
Mesenchymal Stem Cells in Primary Sjögren's Syndrome: Prospective and Challenges. Stem Cells Int 2018; 2018:4357865. [PMID: 30305818 PMCID: PMC6165618 DOI: 10.1155/2018/4357865] [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: 05/25/2018] [Revised: 08/20/2018] [Accepted: 09/02/2018] [Indexed: 02/07/2023] Open
Abstract
Primary Sjögren's syndrome (pSS) is a chronic systemic inflammatory autoimmune disease characterized by lymphocytic infiltrates in exocrine glands. Current approaches do not control harmful autoimmune attacks or prevent irreversible damage and have considerable side effects. Mesenchymal stem cells (MSCs) have been effective in the treatment of several autoimmune diseases. The objective of this review is to illustrate the potential therapeutic role of MSCs in pSS. We summarize the recent advances in what is known about their immunomodulatory function and therapeutic applications in pSS. MSC transfusion can suppress autoimmunity and restore salivary gland secretory function in mouse models and patients with pSS by inducing regulatory T cells, suppressing Th1, Th17, and T follicular helper cell responses. In addition, MSCs can differentiate into salivary epithelial cells, presenting an option as a suitable alternative treatment. We also discuss current bioengineering methods which improve functions of MSCs for pSS. However, there remain many challenges to overcome before their wide clinical application.
Collapse
|
681
|
Burks SR, Nagle ME, Bresler MN, Kim SJ, Star RA, Frank JA. Mesenchymal stromal cell potency to treat acute kidney injury increased by ultrasound-activated interferon-γ/interleukin-10 axis. J Cell Mol Med 2018; 22:6015-6025. [PMID: 30216653 PMCID: PMC6237567 DOI: 10.1111/jcmm.13874] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 07/31/2018] [Accepted: 08/06/2018] [Indexed: 12/26/2022] Open
Abstract
Mesenchymal stromal cell (MSC) therapies combined with renal pulsed focused ultrasound (pFUS) pretreatment increase MSC homing and improve cisplatin-induced acute kidney injury (AKI) better than MSC alone. However, mechanisms underlying improved outcomes remain unknown. We hypothesize pFUS up-regulates renal interferon-γ (IFNγ) and stimulates MSC to produce interleukin-10 (IL-10) after migrating to kidneys. To demonstrate initially, MSC cultured with IFNγ up-regulated IL-10. More MSC-derived IL-10 was detected in kidneys when IFNγ-stimulated MSC were infused and they improved AKI better than unstimulated MSC. Next, IFNγ-knockout mice with AKI received pFUS+MSC, but MSC-derived IL-10 expression and AKI were similar to using MSC alone. AKI in wild-type mice receiving pFUS and IL-10-deficient MSC was also unimproved compared to administering IL-10-deficient MSC alone. Indoleamine 2,3-dioxygenase (IDO), an anti-inflammatory enzyme up-regulated in MSC by IFNγ, was up-regulated during AKI, but was not further elevated in MSC from pFUS-treated kidneys, suggesting that IDO is not involved in improved AKI healing by pFUS+MSC. These data suggest IFNγ is up-regulated by pFUS and after i.v.-infused MSC home to pFUS-treated kidneys, IFNγ stimulates additional IL-10 production by MSC to improve AKI. Analogous mechanisms of ultrasound-treated tissue microenvironments stimulating therapeutic MSC may exist in other pathologies where adjuvant ultrasound techniques are successful.
Collapse
Affiliation(s)
- Scott R Burks
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Matthew E Nagle
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Michele N Bresler
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Saejeong J Kim
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Robert A Star
- Renal Diagnostics and Therapeutics Unit, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Joseph A Frank
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland.,National Institute of Biomedical Imaging and Bioengineering, Bethesda, Maryland
| |
Collapse
|
682
|
Leyendecker A, Pinheiro CCG, Amano MT, Bueno DF. The Use of Human Mesenchymal Stem Cells as Therapeutic Agents for the in vivo Treatment of Immune-Related Diseases: A Systematic Review. Front Immunol 2018; 9:2056. [PMID: 30254638 PMCID: PMC6141714 DOI: 10.3389/fimmu.2018.02056] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/21/2018] [Indexed: 12/13/2022] Open
Abstract
Background: One of the greatest challenges for medicine is to find a safe and effective treatment for immune-related diseases. However, due to the low efficacy of the treatment available and the occurrence of serious adverse effects, many groups are currently searching for alternatives to the traditional therapy. In this regard, the use of human mesenchymal stem cells (hMSCs) represents a great promise for the treatment of a variety of immune-related diseases due to their potent immunomodulatory properties. The main objective of this study is, therefore, to present and summarize, through a systematic review of the literature, in vivo studies in which the efficacy of the administration of hMSCs for the treatment of immune-related diseases was evaluated. Methods: The article search was conducted in PubMed/MEDLINE, Scopus and Web of Science databases. Original research articles assessing the therapeutic potential of hMSCs administration for the in vivo treatment immune-related diseases, published from 1984 to December 2017, were selected and evaluated. Results: A total of 132 manuscripts formed the basis of this systematic review. Most of the studies analyzed reported positive results after hMSCs administration. Clinical effects commonly observed include an increase in the survival rates and a reduction in the severity and incidence of the immune-related diseases studied. In addition, hMSCs administration resulted in an inhibition in the proliferation and activation of CD19+ B cells, CD4+ Th1 and Th17 cells, CD8+ T cells, NK cells, macrophages, monocytes, and neutrophils. The clonal expansion of both Bregs and Tregs cells, however, was stimulated. Administration of hMSCs also resulted in a reduction in the levels of pro-inflammatory cytokines such as IFN-γ, TNF-α, IL-1, IL-2, IL-12, and IL-17 and in an increase in the levels of immunoregulatory cytokines such as IL-4, IL-10, and IL-13. Conclusions: The results obtained in this study open new avenues for the treatment of immune-related diseases through the administration of hMSCs and emphasize the importance of the conduction of further studies in this area.
Collapse
|
683
|
El-Sherbiny YM, El-Jawhari JJ, Moseley TA, McGonagle D, Jones E. T cell immunomodulation by clinically used allogeneic human cancellous bone fragments: a potential novel immunotherapy tool. Sci Rep 2018; 8:13535. [PMID: 30201960 PMCID: PMC6131386 DOI: 10.1038/s41598-018-31979-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 08/17/2018] [Indexed: 01/09/2023] Open
Abstract
Multipotential stromal cells (MSCs) demonstrate strong immunomodulation capabilities following culture expansion. We have previously demonstrated that human cancellous bone fragments (CBFs) clinically used as viable allografts for spinal fusion have resident MSCs that exhibit T cell immunomodulation after monolayer expansion. This study investigated the immunomodulatory ability of these CBFs without MSC culture-expansion. CD4 positive T cells were induced to proliferate using CD3/CD28 stimulation and added to CBFs at different ratios of T cells per gram of CBF. A dose-dependent suppressive effect on T cell proliferation was evident and correlated with increased culture supernatant levels of TGF-ß1, but not PGE2. CBF-driven immunosuppression was reduced in co-cultures with TGF-ß neutralising antibodies and was higher in cell contact compared to non-contact cultures. CBF gene expression profile identified vascular cell adhesion molecule-1, bone marrow stromal antigen 2/CD317 and other interferon signalling pathway members as potential immunomodulatory mediators. The CD317 molecule was detected on the surface of CBF-resident cells confirming the gene expression data. Taken together, these data demonstrate that human clinically used CBFs are inherently immunomodulatory and suggest that these viable allografts may be used to deliver therapeutic immunomodulation for immune-related diseases.
Collapse
Affiliation(s)
- Yasser M El-Sherbiny
- National Institute of Health Research Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK.,Clinical Pathology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt.,Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | - Jehan J El-Jawhari
- National Institute of Health Research Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK.,Clinical Pathology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt.,Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | | | - Dennis McGonagle
- National Institute of Health Research Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK.,Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | - Elena Jones
- National Institute of Health Research Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK. .,Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK.
| |
Collapse
|
684
|
Differences of Clonogenic Mesenchymal Stem Cells on Immunomodulation of Lymphocyte Subsets. J Immunol Res 2018; 2018:7232717. [PMID: 30271793 PMCID: PMC6151204 DOI: 10.1155/2018/7232717] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 08/07/2018] [Accepted: 08/09/2018] [Indexed: 01/14/2023] Open
Abstract
Mesenchymal stem cells (MSC) are a widely used population in cell therapy for their ability to differentiate into distinct tissues and more lately, for their immunomodulatory properties. However, the use of heterogeneous populations could be responsible for the nondesired outcomes reflected in the literature. Here, we analyse the different capacities of five one-cell-derived MSC clones to exert their immunomodulation ex vivo. We assessed proliferation assays in cocultures of MSC clones and purified cluster of differentiation (CD)3+, CD4+, or CD8+ lymphocytes; analysed the regulatory T (Treg) cells fold change rate; determined the effects on viability of peripheral blood mononuclear cells (PBMC); and also measured the coculture cytokine profiles (Th1/Th2). Conditioned media (CM) of different clones were also used to perform both proliferation assays and to analyse Treg fold change. The five clones analysed in this work were able to generate heterogeneous environments. Different clones inhibited proliferation of CD3+ and CD4+ lymphocytes, with different intensities. Surprisingly, all clones promoted proliferation of CD8+ lymphocytes. Different MSC clones and their CM were able to increase the number of Treg with different intensities. Finally, different clones also promoted different effects on the viability of PBMC treated with ultraviolet light. Considering all these data together, it seems that different clones, even from the same donor, can promote a wide spectrum of responses from anti-inflammatory to proinflammatory character. This fact may be important to standardise the design of personalized cell therapy protocols, thus diminishing the aforementioned undesired outcomes existing nowadays in this type of therapies.
Collapse
|
685
|
Hsu FT, Wei ZH, Hsuan YCY, Lin W, Su YC, Liao CH, Hsieh CL. MRI tracking of polyethylene glycol-coated superparamagnetic iron oxide-labelled placenta-derived mesenchymal stem cells toward glioblastoma stem-like cells in a mouse model. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:S448-S459. [PMID: 30198338 DOI: 10.1080/21691401.2018.1499661] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Mesenchymal stem cells (MSCs) that display homing and infiltration properties towards tumor cells are a promising cellular targeting vector for brain tumor therapy but are limited to local-regional delivery in current preclinical models. Here, we investigated whether placenta-derived MSCs (P-MSCs) are a superior cellular vector for systemic targeting of glioblastoma stem-like cells (GSCs), with an imaging modality to real-time monitor the trafficking P-MSCs to glioblastoma sites. Results demonstrated that P-MSCs had greater migratory activity towards GSCs and across blood-brain barrier compared with bone marrow-derived MSCs, and this activity was enhanced by hypoxia precondition. Chemokine ligand 5 was identified as a chemoattractant responsible for the glioblastoma tropism of P-MSCs. Polyethylene glycol-coated superparamagnetic iron oxide (PEG-SPIO) was synthesized for cellular labelling and imaging P-MSCs, displaying high cellular uptake and no cytotoxic effect on P-MSCs cell proliferation or stemness property. The homing effects of intravenously administered PEG-SPIO-labelled P-MSCs towards intracerebral GSCs were able to be detected in mice models through T2-weighted magnetic resonance imaging (MRI). This study suggests the possibility of innovative systemic P-MSC-based cell therapy for aggressive GSCs, developing a state-of-the-art theranostic technique for real-time tracking of therapeutic P-MSCs tumor infiltration through cellular MRI.
Collapse
Affiliation(s)
- Fei-Ting Hsu
- a Department of Radiology , School of Medicine, College of Medicine, Taipei Medical University , Taipei , Taiwan.,b Department of Biological Science and Technology , China Medical University , Taichung , Taiwan.,c Department of Medical Imaging , Taipei Medical University Hospital , Taipei , Taiwan.,d Research Center of Translational Imaging , College of Medicine, Taipei Medical University , Taipei , Taiwan
| | - Zung-Hang Wei
- e Department of Power Mechanical Engineering , National Tsing Hua University , Hsinchu , Taiwan
| | | | - Willie Lin
- f Meridigen Biotech Co., Ltd. , Neihu, Taipei City , Taiwan
| | - Yu-Chin Su
- f Meridigen Biotech Co., Ltd. , Neihu, Taipei City , Taiwan
| | - Chia-Hui Liao
- g The PhD Program for Translational Medicine , College of Medical Science and Technology, Taipei Medical University , Taipei , Taiwan
| | - Chia-Ling Hsieh
- g The PhD Program for Translational Medicine , College of Medical Science and Technology, Taipei Medical University , Taipei , Taiwan.,h Clinical Research Center , Taipei Medical University Hospital, Taipei Medical University , Taipei , Taiwan.,i TMU Research Center of Cancer Translational Medicine , Taipei Medical University , Taipei , Taiwan
| |
Collapse
|
686
|
Pro inflammatory stimuli enhance the immunosuppressive functions of adipose mesenchymal stem cells-derived exosomes. Sci Rep 2018; 8:13325. [PMID: 30190615 PMCID: PMC6127134 DOI: 10.1038/s41598-018-31707-9] [Citation(s) in RCA: 184] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 07/17/2018] [Indexed: 02/07/2023] Open
Abstract
The predominant mechanism by which adipose mesenchymal stem cells (AMSCs) participate to tissue repair is through a paracrine activity and their communication with the inflammatory microenvironment is essential part of this process. This hypothesis has been strengthened by the recent discovery that stem cells release not only soluble factors but also extracellular vesicles, which elicit similar biological activity to the stem cells themselves. We demonstrated that the treatment with inflammatory cytokines increases the immunosuppressive and anti-inflammatory potential of AMSCs-derived exosomes, which acquire the ability to shift macrophages from M1 to M2 phenotype by shuttling miRNA regulating macrophages polarization. This suggests that the immunomodulatory properties of AMSCs-derived exosomes may be not constitutive, but are instead induced by the inflammatory microenvironment.
Collapse
|
687
|
Wang Z, Sun D. Adipose-Derived Mesenchymal Stem Cells: A New Tool for the Treatment of Renal Fibrosis. Stem Cells Dev 2018; 27:1406-1411. [PMID: 30032706 DOI: 10.1089/scd.2017.0304] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
As chronic kidney disease progresses, kidney tissue inevitably undergoes cell loss, accumulation of extracellular matrix, and kidney tissue fibrosis, eventually leading to end-stage renal disease. With the continuous innovation of cell therapy technology, mesenchymal stem cells are used in numerous fields, including cardiovascular diseases, diabetes, and kidney tissue injury repair. Adipose-derived mesenchymal stem cells (AMSCs), a type of pluripotent stem cells, have the potential for self-renewal and proliferation with low immunogenicity and significant anti-inflammatory properties. AMSCs can promote impaired cell regeneration and remodeling in renal lesions, thus avoiding further worsening of renal disease and even blocking or reversing the process of renal fibrosis. In this review, we discuss the mechanisms involved in the treatment of renal fibrosis with AMSCs and summarize the potential hazards that may exist in cell therapy.
Collapse
Affiliation(s)
- Zhuojun Wang
- 1 Department of Nephrology, Affiliated Hospital of Xuzhou Medical University , Xuzhou, China
| | - Dong Sun
- 1 Department of Nephrology, Affiliated Hospital of Xuzhou Medical University , Xuzhou, China .,2 Department of Internal Medicine and Diagnostics, Xuzhou Medical University , Xuzhou, China
| |
Collapse
|
688
|
Current Strategies to Generate Human Mesenchymal Stem Cells In Vitro. Stem Cells Int 2018; 2018:6726185. [PMID: 30224922 PMCID: PMC6129345 DOI: 10.1155/2018/6726185] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/31/2018] [Accepted: 08/09/2018] [Indexed: 12/31/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are heterogeneous multipotent stem cells that are involved in the development of mesenchyme-derived evolving structures and organs during ontogeny. In the adult organism, reservoirs of MSCs can be found in almost all tissues where MSCs contribute to the maintenance of organ integrity. The use of these different MSCs for cell-based therapies has been extensively studied over the past years, which highlights the use of MSCs as a promising option for the treatment of various diseases including autoimmune and cardiovascular disorders. However, the proportion of MSCs contained in primary isolates of adult tissue biopsies is rather low and, thus, vigorous ex vivo expansion is needed especially for therapies that may require extensive and repetitive cell substitution. Therefore, more easily and accessible sources of MSCs are needed. This review summarizes the current knowledge of the different strategies to generate human MSCs in vitro as an alternative method for their applications in regenerative therapy.
Collapse
|
689
|
Park EH, Lim HS, Lee S, Roh K, Seo KW, Kang KS, Shin K. Intravenous Infusion of Umbilical Cord Blood-Derived Mesenchymal Stem Cells in Rheumatoid Arthritis: A Phase Ia Clinical Trial. Stem Cells Transl Med 2018; 7:636-642. [PMID: 30112846 PMCID: PMC6127229 DOI: 10.1002/sctm.18-0031] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 05/04/2018] [Accepted: 05/09/2018] [Indexed: 12/11/2022] Open
Abstract
Based on immunomodulatory actions of human umbilical cord blood‐derived mesenchymal stem cells (hUCB‐MSCs), in vitro or preclinical studies of hUCB‐MSCs have been conducted extensively in rheumatoid arthritis (RA). However, few human trials have investigated the outcomes of hUCB‐MSC infusions. The CURE‐iv trial was a phase I, uncontrolled, open label trial for RA patients with moderate disease activity despite treatment with methotrexate. The patients received a single intravenous infusion of 2.5 × 107, 5 × 107, or 1 × 108 cells of hUCB‐MSCs for 30 minutes, three patients in each cluster, with an increment of cell numbers when there was no dose‐limited adverse event. Clinical and safety assessments were performed during the study period, and serum cytokines were measured at baseline and 24 hours after the infusion. Out of 11 screened RA patients, 9 were enrolled. The participants were predominantly female (78%) and the mean age was 57.4 years. The mean disease duration was 9.5 years, and baseline 28‐joint disease activity score (DAS28; using erythrocyte sedimentation rate) was 4.53. There was no major toxicity in all clusters up to 4 weeks after the infusion. Serum erythrocyte sedimentation rate changes at 4 weeks (n = 9) were −7.9 ± 10.4 (p = .0517) and DAS28 changes were −1.60 ± 1.57 (p = .0159). Reduced levels of IL‐1β, IL‐6, IL‐8, and TNF‐α at 24 hours were observed in the cluster infused with 1 × 108 MSCs. This phase Ia hUCB‐MSC infusion trial for established RA patients revealed no short‐term safety concerns. Stem Cells Translational Medicine2018
Collapse
Affiliation(s)
- Eun Hye Park
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Hee-Suk Lim
- Division of Rheumatology, Department of Internal Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, South Korea
| | - Seunghee Lee
- Institute for Stem cell Regenerative Medicine, Kangstem Biotech, Seoul, South Korea
| | - Kyounghwan Roh
- Institute for Stem cell Regenerative Medicine, Kangstem Biotech, Seoul, South Korea
| | - Kwang-Won Seo
- Institute for Stem cell Regenerative Medicine, Kangstem Biotech, Seoul, South Korea
| | - Kyung-Sun Kang
- Institute for Stem cell Regenerative Medicine, Kangstem Biotech, Seoul, South Korea.,Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Kichul Shin
- Division of Rheumatology, Department of Internal Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, South Korea
| |
Collapse
|
690
|
Chronic Niche Inflammation in Endometriosis-Associated Infertility: Current Understanding and Future Therapeutic Strategies. Int J Mol Sci 2018; 19:ijms19082385. [PMID: 30104541 PMCID: PMC6121292 DOI: 10.3390/ijms19082385] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 08/05/2018] [Accepted: 08/09/2018] [Indexed: 12/13/2022] Open
Abstract
Endometriosis is an estrogen-dependent inflammatory disease that affects up to 10% of women of reproductive age and accounts for up to 50% of female infertility cases. It has been highly associated with poorer outcomes of assisted reproductive technology (ART), including decreased oocyte retrieval, lower implantation, and pregnancy rates. A better understanding of the pathogenesis of endometriosis-associated infertility is crucial for improving infertility treatment outcomes. Current theories regarding how endometriosis reduces fertility include anatomical distortion, ovulatory dysfunction, and niche inflammation-associated peritoneal or implantation defects. This review will survey the latest evidence on the role of inflammatory niche in the peritoneal cavity, ovaries, and uterus of endometriosis patients. Nonhormone treatment strategies that target these inflammation processes are also included. Furthermore, mesenchymal stem cell-based therapies are highlighted for potential endometriosis treatment because of their immunomodulatory effects and tropism toward inflamed lesion foci. Potential applications of stem cell therapy in treatment of endometriosis-associated infertility in particular for safety and efficacy are discussed.
Collapse
|
691
|
Biguetti CC, Vieira AE, Cavalla F, Fonseca AC, Colavite PM, Silva RM, Trombone APF, Garlet GP. CCR2 Contributes to F4/80+ Cells Migration Along Intramembranous Bone Healing in Maxilla, but Its Deficiency Does Not Critically Affect the Healing Outcome. Front Immunol 2018; 9:1804. [PMID: 30147688 PMCID: PMC6095997 DOI: 10.3389/fimmu.2018.01804] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 07/23/2018] [Indexed: 12/29/2022] Open
Abstract
Bone healing depends of a transient inflammatory response, involving selective migration of leukocytes under the control of chemokine system. CCR2 has been regarded as an essential receptor for macrophage recruitment to inflammation and healing sites, but its role in the intramembranous bone healing on craniofacial region remains unknown. Therefore, we investigated the role of CCR2 on F4/80+ cells migration and its consequences to the intramembranous healing outcome. C57BL/6 wild-type (WT) and CCR2KO mice were subjected to upper right incisor extraction, followed by micro-computed tomography, histological, immunological, and molecular analysis along experimental periods. CCR2 was associated with F4/80+ cells influx to the intramembranous bone healing in WT mice, and CCR2+ cells presented a kinetics similar to F4/80+ and CCR5+ cells. By contrast, F4/80+ and CCR5+ cells were significantly reduced in CCR2KO mice. The absence of CCR2 did not cause major microscopic changes in healing parameters, while molecular analysis demonstrated differential genes expression of several molecules between CCR2KO and WT mice. The mRNA expression of TGFB1, RUNX2, and mesenchymal stem cells markers (CXCL12, CD106, OCT4, NANOG, and CD146) was decreased in CCR2KO mice, while IL6, CXCR1, RANKL, and ECM markers (MMP1, 2, 9, and Col1a2) were significantly increased in different periods. Finally, immunofluorescence and FACS revealed that F4/80+ cells are positive for both CCR2 and CCR5, suggesting that CCR5 may account for the remaining migration of the F4/80+ cells in CCR2KO mice. In summary, these results indicate that CCR2+ cells play a primary role in F4/80+ cells migration along healing in intramembranous bones, but its deficiency does not critically impact healing outcome.
Collapse
Affiliation(s)
- Claudia Cristina Biguetti
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, Brazil
| | | | - Franco Cavalla
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, Brazil.,Department of Conservative Dentistry, School of Dentistry, University of Chile, Santiago, Chile
| | - Angélica Cristina Fonseca
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, Brazil
| | - Priscila Maria Colavite
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, Brazil
| | - Renato Menezes Silva
- Department of Endodontics, University of Texas School of Dentistry at Houston, Houston, TX, United States
| | | | | |
Collapse
|
692
|
Bose RJ, Kim BJ, Arai Y, Han IB, Moon JJ, Paulmurugan R, Park H, Lee SH. Bioengineered stem cell membrane functionalized nanocarriers for therapeutic targeting of severe hindlimb ischemia. Biomaterials 2018; 185:360-370. [PMID: 30273835 DOI: 10.1016/j.biomaterials.2018.08.018] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 07/31/2018] [Accepted: 08/06/2018] [Indexed: 01/15/2023]
Abstract
Bioengineering strategies to enhance the natural targeting function of nanocarriers would expand their therapeutic applications. Here, we designed bioengineered stem cell membrane-functionalized nanocarriers (BSMNCs) harboring C-X-C chemokine receptor type 4 (CXCR4) to achieve robust targeting and also to increase their retention time in ischemic tissue. Stem cell membrane coated nanocarrier (SMNCs) or poly (lactic-co-glycolic acid) (PLGA) nanocarriers (PNCs) and BSMNCs were prepared by functionalizing PNCs with human adipose-derived stem cells (hASCs) membranes and hASCs engineered to overexpress CXCR4-receptor, respectively. The functionalization of PNCs with stem cell membranes derived from hASCs significantly enhance the nanocarrier penetration across endothelial cell barrier compare to PNCs. In addition, stem cell membrane functionalization on PNCs also significantly decreased the nanoparticles uptake in J774 (murine) and THP (human) macrophages respectively from 84% to 76%-29% and 24%. Interestingly, BSMNCs showed much higher level of accumulation in ischemic tissue than SMNCs. Systemic retro-orbital injection of BSMNCs loaded with VEGF into mice with hindlimb ischemia resulted substantially enhancement of blood reperfusion, muscle repair, and limb salvage compared to animals treated with SMNCs loaded with similar concentration of VEGF. The reported strategy could be used to create biocompatible and custom-tailored biomimetic nanoparticles with various hybrid functionalities, which may overcome the limitations of current nanoparticle-based therapeutic and imaging platforms.
Collapse
Affiliation(s)
- Rajendran Jc Bose
- Advanced Biomaterials and Stem Cell Engineering Laboratory, School of Integrative Engineering, Chung-Ang University, 84 Heuk Seok-Ro, Dongjak-Gu, Seoul, South Korea; Stem Cell Engineering Laboratory, Department of Biomedical Science, CHA University, Pangyo-Ro 335, Bundang-gu, Seongnam-si, Gyeonggi-do 463-400, South Korea; Molecular Imaging Program at Stanford (MIPS), and Bio-X Program, Department of Radiology, School of Medicine, Stanford University, Stanford, CA, 94305-5427, USA; Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Stanford University, CA, 94305-5427, USA
| | - Byoung Ju Kim
- Stem Cell Engineering Laboratory, Department of Biomedical Science, CHA University, Pangyo-Ro 335, Bundang-gu, Seongnam-si, Gyeonggi-do 463-400, South Korea; Stem Cells Integrative Engineering Laboratory, Department of Medical Biotechnology, Dongguk University, 32 Dongguk-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do, 10326, South Korea
| | - Yoshie Arai
- Stem Cell Engineering Laboratory, Department of Biomedical Science, CHA University, Pangyo-Ro 335, Bundang-gu, Seongnam-si, Gyeonggi-do 463-400, South Korea; Stem Cells Integrative Engineering Laboratory, Department of Medical Biotechnology, Dongguk University, 32 Dongguk-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do, 10326, South Korea
| | - In-Bo Han
- Department of Neurosurgery, CHA University, Pangyo-Ro 335, Bundang-gu, Seongnam-si, Gyeonggi-do 463-400, South Korea
| | - James J Moon
- Department of Pharmaceutical Sciences, Department of Biomedical Engineering & Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ramasamy Paulmurugan
- Molecular Imaging Program at Stanford (MIPS), and Bio-X Program, Department of Radiology, School of Medicine, Stanford University, Stanford, CA, 94305-5427, USA; Canary Center at Stanford for Cancer Early Detection, Department of Radiology, School of Medicine, Stanford University, CA, 94305-5427, USA
| | - Hansoo Park
- Advanced Biomaterials and Stem Cell Engineering Laboratory, School of Integrative Engineering, Chung-Ang University, 84 Heuk Seok-Ro, Dongjak-Gu, Seoul, South Korea.
| | - Soo-Hong Lee
- Stem Cell Engineering Laboratory, Department of Biomedical Science, CHA University, Pangyo-Ro 335, Bundang-gu, Seongnam-si, Gyeonggi-do 463-400, South Korea; Stem Cells Integrative Engineering Laboratory, Department of Medical Biotechnology, Dongguk University, 32 Dongguk-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do, 10326, South Korea.
| |
Collapse
|
693
|
Gaber T, Schönbeck K, Hoff H, Tran CL, Strehl C, Lang A, Ohrndorf S, Pfeiffenberger M, Röhner E, Matziolis G, Burmester GR, Buttgereit F, Hoff P. CTLA-4 Mediates Inhibitory Function of Mesenchymal Stem/Stromal Cells. Int J Mol Sci 2018; 19:ijms19082312. [PMID: 30087255 PMCID: PMC6121442 DOI: 10.3390/ijms19082312] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 08/01/2018] [Accepted: 08/02/2018] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are stem cells of the connective tissue, possess a plastic phenotype, and are able to differentiate into various tissues. Besides their role in tissue regeneration, MSCs perform additional functions as a modulator or inhibitor of immune responses. Due to their pleiotropic function, MSCs have also gained therapeutic importance for the treatment of autoimmune diseases and for improving fracture healing and cartilage regeneration. However, the therapeutic/immunomodulatory mode of action of MSCs is largely unknown. Here, we describe that MSCs express the inhibitory receptor CTLA-4 (cytotoxic T lymphocyte antigen 4). We show that depending on the environmental conditions, MSCs express different isoforms of CTLA-4 with the secreted isoform (sCTLA-4) being the most abundant under hypoxic conditions. Furthermore, we demonstrate that the immunosuppressive function of MSCs is mediated mainly by the secretion of CTLA-4. These findings open new ways for treatment when tissue regeneration/fracture healing is difficult.
Collapse
Affiliation(s)
- Timo Gaber
- Department of Rheumatology and Clinical Immunology, Charité⁻Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.
- German Rheumatism Research Centre (DRFZ) Berlin, A Leibniz Institute, 10117 Berlin, Germany.
| | - Kerstin Schönbeck
- Department of Rheumatology and Clinical Immunology, Charité⁻Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.
- German Rheumatism Research Centre (DRFZ) Berlin, A Leibniz Institute, 10117 Berlin, Germany.
| | - Holger Hoff
- German Rheumatism Research Centre (DRFZ) Berlin, A Leibniz Institute, 10117 Berlin, Germany.
| | - Cam Loan Tran
- Department of Rheumatology and Clinical Immunology, Charité⁻Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.
- German Rheumatism Research Centre (DRFZ) Berlin, A Leibniz Institute, 10117 Berlin, Germany.
| | - Cindy Strehl
- Department of Rheumatology and Clinical Immunology, Charité⁻Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.
- German Rheumatism Research Centre (DRFZ) Berlin, A Leibniz Institute, 10117 Berlin, Germany.
| | - Annemarie Lang
- Department of Rheumatology and Clinical Immunology, Charité⁻Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.
- German Rheumatism Research Centre (DRFZ) Berlin, A Leibniz Institute, 10117 Berlin, Germany.
| | - Sarah Ohrndorf
- Department of Rheumatology and Clinical Immunology, Charité⁻Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.
| | - Moritz Pfeiffenberger
- Department of Rheumatology and Clinical Immunology, Charité⁻Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.
- German Rheumatism Research Centre (DRFZ) Berlin, A Leibniz Institute, 10117 Berlin, Germany.
| | - Eric Röhner
- Department of Orthopedics, Campus Eisenberg, Jena University Hospital, Klosterlausnitzer Straße 81, 07607 Eisenberg, Germany.
| | - Georg Matziolis
- Department of Orthopedics, Campus Eisenberg, Jena University Hospital, Klosterlausnitzer Straße 81, 07607 Eisenberg, Germany.
| | - Gerd-R Burmester
- Department of Rheumatology and Clinical Immunology, Charité⁻Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.
- German Rheumatism Research Centre (DRFZ) Berlin, A Leibniz Institute, 10117 Berlin, Germany.
| | - Frank Buttgereit
- Department of Rheumatology and Clinical Immunology, Charité⁻Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.
- German Rheumatism Research Centre (DRFZ) Berlin, A Leibniz Institute, 10117 Berlin, Germany.
| | - Paula Hoff
- Department of Rheumatology and Clinical Immunology, Charité⁻Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany.
- German Rheumatism Research Centre (DRFZ) Berlin, A Leibniz Institute, 10117 Berlin, Germany.
- Department of Orthopedics, Campus Eisenberg, Jena University Hospital, Klosterlausnitzer Straße 81, 07607 Eisenberg, Germany.
- Endokrinologikum Berlin, 10117 Berlin, Germany.
| |
Collapse
|
694
|
Bone Morphogenetic Protein 6 Inhibits the Immunomodulatory Property of BMMSCs via Id1 in Sjögren's Syndrome. Stem Cells Int 2018; 2018:9837035. [PMID: 30174696 PMCID: PMC6098892 DOI: 10.1155/2018/9837035] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 06/19/2018] [Indexed: 12/29/2022] Open
Abstract
Mesenchymal stem cells (MSCs) treatment has emerged as a promising approach for treating Sjögren's syndrome (SS). Impaired immunoregulatory activities of bone marrow mesenchymal stem cells (BMMSCs) are found in both SS patients and animal models, and the underlying mechanism is poorly understood. Increased expression of BMP6 is reported to be related to SS. The aim herein was to determine the effects of BMP6 on BMMSCs function. BMMSCs were isolated from SS patients and NOD mice and showed a high level of BMP6 expression. The effects of BMP6 on BMMSCs function were investigated using in vitro BMMSCs differentiation and in vitro and in vivo T cell proliferation and polarization assays. BMP6 increased osteogenic differentiation of BMMSCs and inhibited the immunomodulatory properties of BMMSCs. BMP6 enhanced T cell proliferation and Th1/Th17 differentiation in a T cell-BMMSC coculture system. Mechanistically, BMP6 downregulated PGE2 and upregulated IFN-gamma via Id1 (inhibitor of DNA-binding protein 1). Neutralizing BMP6 and knockdown of Id1 could restore the BMMSCs immunosuppressive function both in vitro and in vivo. The present results suggest a novel role of Id1 in BMP-mediated MSCs function, which may contribute to a better understanding of the mechanism of action of MSCs in treating autoimmune diseases.
Collapse
|
695
|
Tasnim N, De la Vega L, Anil Kumar S, Abelseth L, Alonzo M, Amereh M, Joddar B, Willerth SM. 3D Bioprinting Stem Cell Derived Tissues. Cell Mol Bioeng 2018; 11:219-240. [PMID: 31719887 PMCID: PMC6816617 DOI: 10.1007/s12195-018-0530-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 05/14/2018] [Indexed: 12/11/2022] Open
Abstract
Stem cells offer tremendous promise for regenerative medicine as they can become a variety of cell types. They also continuously proliferate, providing a renewable source of cells. Recently, it has been found that 3D printing constructs using stem cells, can generate models representing healthy or diseased tissues, as well as substitutes for diseased and damaged tissues. Here, we review the current state of the field of 3D printing stem cell derived tissues. First, we cover 3D printing technologies and discuss the different types of stem cells used for tissue engineering applications. We then detail the properties required for the bioinks used when printing viable tissues from stem cells. We give relevant examples of such bioprinted tissues, including adipose tissue, blood vessels, bone, cardiac tissue, cartilage, heart valves, liver, muscle, neural tissue, and pancreas. Finally, we provide future directions for improving the current technologies, along with areas of focus for future work to translate these exciting technologies into clinical applications.
Collapse
Affiliation(s)
- Nishat Tasnim
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, 500 W University Avenue, El Paso, TX 79968 USA
- Border Biomedical Research Center, University of Texas at El Paso, 500 W University Avenue, El Paso, TX 79968 USA
| | - Laura De la Vega
- Department of Mechanical Engineering, University of Victoria, 3800 Finnerty Road, Victoria, BC V8W 2Y2 Canada
- Border Biomedical Research Center, University of Texas at El Paso, 500 W University Avenue, El Paso, TX 79968 USA
| | - Shweta Anil Kumar
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, 500 W University Avenue, El Paso, TX 79968 USA
- Border Biomedical Research Center, University of Texas at El Paso, 500 W University Avenue, El Paso, TX 79968 USA
| | - Laila Abelseth
- Biomedical Engineering Program, University of Victoria, 3800 Finnerty Road, Victoria, BC V8W 2Y2 Canada
- Border Biomedical Research Center, University of Texas at El Paso, 500 W University Avenue, El Paso, TX 79968 USA
| | - Matthew Alonzo
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, 500 W University Avenue, El Paso, TX 79968 USA
- Border Biomedical Research Center, University of Texas at El Paso, 500 W University Avenue, El Paso, TX 79968 USA
| | - Meitham Amereh
- Faculty of Engineering, University of British Columbia-Okanagan Campus, Kelowna, Canada
- Border Biomedical Research Center, University of Texas at El Paso, 500 W University Avenue, El Paso, TX 79968 USA
| | - Binata Joddar
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, 500 W University Avenue, El Paso, TX 79968 USA
- Border Biomedical Research Center, University of Texas at El Paso, 500 W University Avenue, El Paso, TX 79968 USA
- Division of Medical Sciences, University of Victoria, 3800 Finnerty Road, Victoria, BC V8W 2Y2 Canada
| | - Stephanie M. Willerth
- Biomedical Engineering Program, University of Victoria, 3800 Finnerty Road, Victoria, BC V8W 2Y2 Canada
- Border Biomedical Research Center, University of Texas at El Paso, 500 W University Avenue, El Paso, TX 79968 USA
- Division of Medical Sciences, University of Victoria, 3800 Finnerty Road, Victoria, BC V8W 2Y2 Canada
- International Collaboration on Repair Discoveries, University of British Columbia, 818 West 10th Avenue, Vancouver, BC V5Z 1M9 Canada
| |
Collapse
|
696
|
Nunes HC, Scarano WR, Deffune E, Felisbino SL, Porreca I, Delella FK. Bisphenol a and mesenchymal stem cells: Recent insights. Life Sci 2018; 206:22-28. [DOI: 10.1016/j.lfs.2018.05.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 05/07/2018] [Accepted: 05/09/2018] [Indexed: 02/08/2023]
|
697
|
Nugraha AP, Narmada IB, Ernawati DS, Dinaryanti A, Hendrianto E, Ihsan IS, Riawan W, Rantam FA. Osteogenic potential of gingival stromal progenitor cells cultured in platelet rich fibrin is predicted by core-binding factor subunit-α1/Sox9 expression ratio ( in vitro). F1000Res 2018; 7:1134. [PMID: 30430007 PMCID: PMC6097418 DOI: 10.12688/f1000research.15423.1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/10/2018] [Indexed: 12/16/2022] Open
Abstract
Background: Alveolar bone defect regeneration has long been problematic in the field of dentistry. Gingival stromal progenitor cells (GSPCs) offer a promising solution for alveolar bone regeneration. In order to optimally differentiate and proliferate progenitor cells, growth factors (GFs) are required. Platelet rich fibrin (PRF) has many GFs and can be easily manufactured. Core-binding factor subunit-α1 (CBF-α1) constitutes a well-known osteogenic differentiation transcription factor in SPCs. Sox9, as a chondrogenic transcription factor, interacts and inhibits CBF-α1, but its precise role in direct in vitro osteogenesis remains unknown. GSPCs cultured in vitro in PRF to optimally stimulate osteogenic differentiation has been largely overlooked. The aim of this study was to analyze GSPCs cultured in PRF osteogenic differentiation predicted by CBF-α1/Sox9. Methods: This study used a true experimental with post-test only control group design and random sampling. GPSCs isolated from the lower gingiva of four healthy, 250-gram, 1-month old, male Wistar rats ( Rattus Novergicus) were cultured for two weeks, passaged every 4-5 days. GSPCs in passage 3-5 were cultured in five M24 plates (N=108; n=6/group) for Day 7, Day 14, and Day 21 in three different mediums (control negative group: αModified Eagle Medium; control positive group: High Glucose-Dulbecco's Modified Eagle Medium (DMEM-HG) + osteogenic medium; Treatment group: DMEM-HG + osteogenic medium + PRF). CBF-α1 and Sox9 were examined with ICC monoclonal antibody. A one-way ANOVA continued with Tukey HSD test (p<0.05) based on Kolmogorov-Smirnov and Levene's tests (p>0.05) was performed. Results: The treatment group showed the highest CBF-α1/Sox9 ratio (16.00±3.000/14.33±2.517) on Day 7, while the lowest CBF-α1/Sox9 ratio (3.33±1.528/3.67±1.155) occurred in the control negative group on Day 21, with significant difference between the groups (p<0.05). Conclusion: GSPCs cultured in PRF had potential osteogenic differentiation ability predicted by the CBF-α1/sox9 ratio.
Collapse
Affiliation(s)
- Alexander Patera Nugraha
- Graduate School of Immunology, Postgraduate School, Universitas Airlangga, Surabaya, 60132, Indonesia
- Orthodontic Department, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, 60132, Indonesia
- Stem Cell Research and Development Center, Universitas Airlangga, Surabaya, 60132, Indonesia
| | - Ida Bagus Narmada
- Orthodontic Department, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, 60132, Indonesia
| | - Diah Savitri Ernawati
- Oral Medicine Department, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, 60132, Indonesia
| | - Aristika Dinaryanti
- Stem Cell Research and Development Center, Universitas Airlangga, Surabaya, 60132, Indonesia
| | - Eryk Hendrianto
- Stem Cell Research and Development Center, Universitas Airlangga, Surabaya, 60132, Indonesia
| | - Igo Syaiful Ihsan
- Stem Cell Research and Development Center, Universitas Airlangga, Surabaya, 60132, Indonesia
| | - Wibi Riawan
- Biochemistry Biomolecular Laboratory, Faculty of Medicine, Universitas Brawijaya, Malang, 65145, Indonesia
| | - Fedik Abdul Rantam
- Stem Cell Research and Development Center, Universitas Airlangga, Surabaya, 60132, Indonesia
- Virology and Immunology Laboratory, Microbiology Department, Faculty of Veterinary Medicine, Universitas Airlangga., Surabaya, 60132, Indonesia
| |
Collapse
|
698
|
Bunpetch V, Wu H, Zhang S, Ouyang H. From "Bench to Bedside": Current Advancement on Large-Scale Production of Mesenchymal Stem Cells. Stem Cells Dev 2018; 26:1662-1673. [PMID: 28934885 DOI: 10.1089/scd.2017.0104] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are the primary cell source in cell therapy and regenerative medicine due to its extraordinary self-renewing capacity and multilineage differentiation potential. Clinical trials involving MSCs are being conducted in a range of human diseases and the number of registered cases is continuously increasing. However, a wide gap exists between the number of MSCs obtainable from the donor site and the number required for implantation to damage tissues, and also between MSC scalability and MSC phenotype stability. The clinical translation of MSCs necessitates a scalable expansion bioprocess for the biomanufacturing of therapeutically qualified cells. This review presents current achievements for expansion of MSCs. Issues involving culture condition modification, bioreactor systems, as well as microcarrier and scaffold platforms for optimal MSC systems are discussed. Most importantly, the gap between current MSC expansion and clinical application, as well as outbreak directions for the future are discussed. The present systemic review will bring new insights into future large-scale MSC expansion and clinical application.
Collapse
Affiliation(s)
- Varitsara Bunpetch
- 1 Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University , Hangzhou, China .,2 Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou, China .,3 Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University , Hangzhou, China
| | - Haoyu Wu
- 1 Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University , Hangzhou, China .,2 Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou, China .,3 Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University , Hangzhou, China
| | - Shufang Zhang
- 1 Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University , Hangzhou, China .,2 Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou, China .,3 Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University , Hangzhou, China
| | - Hongwei Ouyang
- 1 Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University , Hangzhou, China .,2 Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou, China .,3 Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University , Hangzhou, China .,4 State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University , Hangzhou, China .,5 Department of Sports Medicine, School of Medicine, Zhejiang University , Hangzhou, China
| |
Collapse
|
699
|
Bianco ST, Moser HL, Galatz LM, Huang AH. Biologics and stem cell-based therapies for rotator cuff repair. Ann N Y Acad Sci 2018; 1442:35-47. [PMID: 30008172 DOI: 10.1111/nyas.13918] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 06/01/2018] [Accepted: 06/11/2018] [Indexed: 12/20/2022]
Abstract
The rotator cuff is composed of several distinct muscles and tendons that function in concert to coordinate shoulder motion. Injuries to these tendons frequently result in permanent dysfunction and persistent pain. Despite considerable advances in operation techniques, surgical repair alone still does not fully restore rotator cuff function. This review focuses on recent research in the use of biologics and stem cell-based therapies to augment repair, highlighting promising avenues for future work and remaining challenges. While a number of animal models are used for rotator cuff studies, the anatomy of the rotator cuff varies dramatically between species. Since the rodent rotator cuff shares the most anatomical features with the human, this review will focus primarily on rodent models to enable consistent interpretation of outcome measures.
Collapse
Affiliation(s)
- Spencer T Bianco
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Helen L Moser
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, New York.,Shoulder, Elbow and Orthopaedic Sports Medicine, Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Leesa M Galatz
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alice H Huang
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
700
|
Langrzyk A, Nowak WN, Stępniewski J, Jaźwa A, Florczyk-Soluch U, Józkowicz A, Dulak J. Critical View on Mesenchymal Stromal Cells in Regenerative Medicine. Antioxid Redox Signal 2018; 29:169-190. [PMID: 28874054 DOI: 10.1089/ars.2017.7159] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE The belief in the potency of stem cells has resulted in the medical applications of numerous cell types for organ repair, often with the low adherence to methodological stringency. Such uncritical enthusiasm is mainly presented in the approaches employing so-called mesenchymal stem cells (MSC), for the treatment of numerous, unrelated conditions. However, it should be stressed that such broad clinical applications of MSC are mostly based on the belief that MSC can efficiently differentiate into multiple cell types, not only osteoblasts, chondrocytes and adipose cells. Recent Advances: Studies employing lineage tracing established more promising markers to characterize MSC identity and localization in vivo and confirmed the differences between MSC isolated from various organs. Furthermore, preclinical and clinical experiments proved that transdifferentiation of MSC is unlikely to contribute to repair of numerous tissues, including the heart. Therefore, the salvage hypotheses, like MSC fusion with cells in target organs or the paracrine mechanisms, were proposed to justify the widespread application of MSC and to explain transient, if any, effects. CRITICAL ISSUES The lack of standardization concerning the cells markers, their origin and particularly the absence of stringent functional characterization of MSC, leads to propagation of the worrying hype despite the lack of convincing therapeutic efficiency of MSC. FUTURE DIRECTIONS The adherence to rigorous methodological rules is necessary to prevent the application of procedures which can be dangerous for patients and scientific research on the medical application of stem cells. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
| | - Witold N Nowak
- 2 Cardiovascular Division, King's College London , London, United Kingdom .,3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Jacek Stępniewski
- 3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Agnieszka Jaźwa
- 3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Urszula Florczyk-Soluch
- 3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Alicja Józkowicz
- 3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| | - Józef Dulak
- 1 Kardio-Med Silesia , Zabrze, Poland .,3 Department of Medical Biotechnology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University , Kraków, Poland
| |
Collapse
|