651
|
Norozi F, Ahmadzadeh A, Shahrabi S, Vosoughi T, Saki N. Mesenchymal stem cells as a double-edged sword in suppression or progression of solid tumor cells. Tumour Biol 2016; 37:11679-11689. [PMID: 27440203 DOI: 10.1007/s13277-016-5187-7] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 07/13/2016] [Indexed: 02/07/2023] Open
Abstract
Tumor cells are able to attract mesenchymal stem cells (MSCs) to primary tumor site. On the other hand, MSCs secrete various factors to attract tumor cells towards BM. In this review, in addition to assessment of MSCs function at tumor sites and their impact on growth and metastasis of tumor cells, the importance of MSC in attraction of malignant cells to BM and their involvement in drug resistance of tumor cells have also been studied. Relevant literature was identified by a PubMed search (2000-2015) of English-language literature using the terms mesenchymal stem cells, cancer cell, metastasis, and tumor microenvironment. MSCs migrate towards tumor microenvironment and are involved in both pro-tumorigenic and antitumorigenic functions. The dual function of MSCs at tumor sites is dependent upon a variety of factors, including the type and origin of MSCs, the cancer cell line under study, in vivo or in vitro conditions, the factors secreted by MSCs and interactions between MSCs, host immune cells and cancer cells. Therefore, MSCs can be regarded both as friends and enemies of cancer cells. Although the role of a number of pathways, including IL-6/STAT3 pathway, has been indicated in controlling the interaction between MSCs and tumor cells, other mechanisms by which MSCs can control the tumor cells are not clear yet. A better understanding of these mechanisms through further studies can determine the exact role of MSCs in cancer progression and identify them as important therapeutic agents or targets.
Collapse
Affiliation(s)
- Fatemeh Norozi
- Health Research Institute, Research Center of Thalassemia & Hemoglobinopathy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ahmad Ahmadzadeh
- Health Research Institute, Research Center of Thalassemia & Hemoglobinopathy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Saeid Shahrabi
- Department of biochemistry and hematology, Faculty of Medicine, Semnan University of medical sciences, Semnan, Iran
| | - Tina Vosoughi
- Health Research Institute, Research Center of Thalassemia & Hemoglobinopathy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Najmaldin Saki
- Health Research Institute, Research Center of Thalassemia & Hemoglobinopathy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
652
|
Mesenchymal stem cells attenuate inflammatory processes in the heart and lung via inhibition of TNF signaling. Basic Res Cardiol 2016; 111:54. [PMID: 27435289 PMCID: PMC4951509 DOI: 10.1007/s00395-016-0573-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 07/13/2016] [Indexed: 12/24/2022]
Abstract
Mesenchymal stem cells (MSC) have been used to treat different clinical conditions although the mechanisms by which pathogenetic processes are affected are still poorly understood. We have previously analyzed the homing of bone marrow-derived MSC to diseased tissues characterized by a high degree of mononuclear cell infiltration and postulated that MSC might modulate inflammatory responses. Here, we demonstrate that MSC mitigate adverse tissue remodeling, improve organ function, and extend lifespan in a mouse model of inflammatory dilative cardiomyopathy (DCM). Furthermore, MSC attenuate Lipopolysaccharide-induced acute lung injury indicating a general role in the suppression of inflammatory processes. We found that MSC released sTNF-RI, which suppressed activation of the NFκBp65 pathway in cardiomyocytes during DCM in vivo. Substitution of MSC by recombinant soluble TNF-R partially recapitulated the beneficial effects of MSC while knockdown of TNF-R prevented MSC-mediated suppression of the NFκBp65 pathway and improvement of tissue pathology. We conclude that sTNF-RI is a major part of the paracrine machinery by which MSC effect local inflammatory reactions.
Collapse
|
653
|
Human Cardiac Mesenchymal Stromal Cells with CD105+CD34- Phenotype Enhance the Function of Post-Infarction Heart in Mice. PLoS One 2016; 11:e0158745. [PMID: 27415778 PMCID: PMC4945149 DOI: 10.1371/journal.pone.0158745] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 06/21/2016] [Indexed: 01/01/2023] Open
Abstract
Aims The aim of the present study was to isolate mesenchymal stromal cells (MSC) with CD105+CD34- phenotype from human hearts, and to investigate their therapeutic potential in a mouse model of hindlimb ischemia and myocardial infarction (MI). The study aimed also to investigate the feasibility of xenogeneic MSCs implantation. Methods and Results MSC isolated from human hearts were multipotent cells. Separation of MSC with CD105+CD34- phenotype limited the heterogeneity of the originally isolated cell population. MSC secreted a number of anti-inflammatory and proangiogenic cytokines (mainly IL-6, IL-8, and GRO). Human MSC were transplanted into C57Bl/6NCrl mice. Using the mouse model of hindlimb ischemia it was shown that human MSC treated mice demonstrated a higher capillary density 14 days after injury. It was also presented that MSC administrated into the ischemic muscle facilitated fast wound healing (functional recovery by ischemic limb). MSC transplanted into an infarcted myocardium reduced the post-infarction scar, fibrosis, and increased the number of blood vessels both in the border area, and within the post-infarction scar. The improvement of left ventricular ejection fraction was also observed. Conclusion In two murine models (hindlimb ischemia and MI) we did not observe the xenotransplant rejection. Indeed, we have shown that human cardiac mesenchymal stromal cells with CD105+CD34- phenotype exhibit therapeutic potential. It seems that M2 macrophages are essential for healing and repair of the post-infarcted heart.
Collapse
|
654
|
de Lima KA, de Oliveira GLV, Yaochite JNU, Pinheiro DG, de Azevedo JTC, Silva WA, Covas DT, Couri CEB, Simões BP, Voltarelli JC, Oliveira MC, Malmegrim KCR. Transcriptional profiling reveals intrinsic mRNA alterations in multipotent mesenchymal stromal cells isolated from bone marrow of newly-diagnosed type 1 diabetes patients. Stem Cell Res Ther 2016; 7:92. [PMID: 27406064 PMCID: PMC4942931 DOI: 10.1186/s13287-016-0351-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 04/12/2016] [Accepted: 06/22/2016] [Indexed: 02/07/2023] Open
Abstract
Background Bone marrow multipotent mesenchymal stromal cells (MSCs) are a diverse subset of precursors that contribute to the homeostasis of the hematopoietic niche. MSCs can be isolated and expanded in vitro and have unique immunomodulatory and regenerative properties that make them attractive for the treatment of autoimmune diseases, including type 1 diabetes (T1D). Whether autologous or allogeneic MSCs are more suitable for therapeutic purposes has not yet been established. While autologous MSCs may present abnormal function, allogeneic cells may be recognized and rejected by the host immune system. Thus, studies that investigate biological characteristics of MSCs isolated from T1D patients are essential to guide future clinical applications. Methods Bone marrow-derived MSCs from recently diagnosed type 1 diabetes patients (T1D-MSCs) were compared with those from healthy individuals (C-MSCs) for morphological and immunophenotypic characteristics and for differentiation potential. Bioinformatics approaches allowed us to match absolute and differential gene expression of several adhesion molecules, immune mediators, growth factors, and their receptors involved with hematopoietic support and immunomodulatory properties of MSCs. Finally, the differentially expressed genes were collated for functional pathway enrichment analysis. Results T1D-MSCs and C-MSCs were similar for morphology, immunophenotype, and differentiation potential. Our absolute gene expression results supported previous literature reports, while also detecting new potential molecules related to bone marrow-derived MSC functions. T1D-MSCs showed intrinsic abnormalities in mRNA expression, including the immunomodulatory molecules VCAM-1, CXCL12, HGF, and CCL2. Pathway analyses revealed activation of sympathetic nervous system and JAK STAT signaling in T1D-MSCs. Conclusions Collectively, our results indicate that MSCs isolated from T1D patients present intrinsic transcriptional alterations that may affect their therapeutic potential. However, the implications of these abnormalities in T1D development as well as in the therapeutic efficacy of autologous MSCs require further investigation. Electronic supplementary material The online version of this article (doi:10.1186/s13287-016-0351-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kalil A de Lima
- Center for Cell-Based Research, Regional Blood Center of Ribeirao Preto, Ribeirao Preto Medical, University of Sao Paulo, Ribeirao Preto, Brazil. .,Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil. .,, Tenente Catao Roxo, 2501, Monte Alegre, 14051-140, Ribeirao Preto, Sao Paulo, Brazil.
| | - Gislane L V de Oliveira
- Center for Cell-Based Research, Regional Blood Center of Ribeirao Preto, Ribeirao Preto Medical, University of Sao Paulo, Ribeirao Preto, Brazil.,Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Juliana N U Yaochite
- Center for Cell-Based Research, Regional Blood Center of Ribeirao Preto, Ribeirao Preto Medical, University of Sao Paulo, Ribeirao Preto, Brazil.,Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil.,Department of Clinical and Toxicological Analysis, Federal University of Ceará, Fortaleza, Ceara, Brazil
| | - Daniel G Pinheiro
- Center for Cell-Based Research, Regional Blood Center of Ribeirao Preto, Ribeirao Preto Medical, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Júlia T C de Azevedo
- Center for Cell-Based Research, Regional Blood Center of Ribeirao Preto, Ribeirao Preto Medical, University of Sao Paulo, Ribeirao Preto, Brazil.,Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Wilson Araujo Silva
- Center for Cell-Based Research, Regional Blood Center of Ribeirao Preto, Ribeirao Preto Medical, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Dimas T Covas
- Center for Cell-Based Research, Regional Blood Center of Ribeirao Preto, Ribeirao Preto Medical, University of Sao Paulo, Ribeirao Preto, Brazil.,Department of Clinical Medicine, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Carlos E B Couri
- Center for Cell-Based Research, Regional Blood Center of Ribeirao Preto, Ribeirao Preto Medical, University of Sao Paulo, Ribeirao Preto, Brazil.,Department of Clinical Medicine, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Belinda P Simões
- Center for Cell-Based Research, Regional Blood Center of Ribeirao Preto, Ribeirao Preto Medical, University of Sao Paulo, Ribeirao Preto, Brazil.,Department of Clinical Medicine, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Julio C Voltarelli
- Center for Cell-Based Research, Regional Blood Center of Ribeirao Preto, Ribeirao Preto Medical, University of Sao Paulo, Ribeirao Preto, Brazil.,Department of Clinical Medicine, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Maria C Oliveira
- Center for Cell-Based Research, Regional Blood Center of Ribeirao Preto, Ribeirao Preto Medical, University of Sao Paulo, Ribeirao Preto, Brazil.,Department of Clinical Medicine, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Kelen C R Malmegrim
- Center for Cell-Based Research, Regional Blood Center of Ribeirao Preto, Ribeirao Preto Medical, University of Sao Paulo, Ribeirao Preto, Brazil.,Department of Clinical, Toxicological and Bromatological Analysis, Faculty of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, Brazil
| |
Collapse
|
655
|
Intravenous administration of bone marrow-derived multipotent mesenchymal stromal cells enhances the recruitment of CD11b+ myeloid cells to the lungs and facilitates B16-F10 melanoma colonization. Exp Cell Res 2016; 345:141-9. [DOI: 10.1016/j.yexcr.2015.05.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Revised: 05/25/2015] [Accepted: 05/26/2015] [Indexed: 01/01/2023]
|
656
|
Skardal A, Murphy SV, Crowell K, Mack D, Atala A, Soker S. A tunable hydrogel system for long-term release of cell-secreted cytokines and bioprinted in situ wound cell delivery. J Biomed Mater Res B Appl Biomater 2016; 105:1986-2000. [PMID: 27351939 DOI: 10.1002/jbm.b.33736] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 05/18/2016] [Accepted: 06/06/2016] [Indexed: 12/21/2022]
Abstract
For many cellular therapies being evaluated in preclinical and clinical trials, the mechanisms behind their therapeutic effects appear to be the secretion of growth factors and cytokines, also known as paracrine activity. Often, delivered cells are transient, and half-lives of the growth factors that they secrete are short, limiting their long-term effectiveness. The goal of this study was to optimize a hydrogel system capable of in situ cell delivery that could sequester and release growth factors secreted from those cells after the cells were no longer present. Here, we demonstrate the use of a fast photocross-linkable heparin-conjugated hyaluronic acid (HA-HP) hydrogel as a cell delivery vehicle for sustained growth factor release, which extends paracrine activity. The hydrogel could be modulated through cross-linking geometries and heparinization to support sustained release proteins and heparin-binding growth factors. To test the hydrogel in vivo, we used it to deliver amniotic fluid-derived stem (AFS) cells, which are known to secrete cytokines and growth factors, in full thickness skin wounds in a nu/nu murine model. Despite transience of the AFS cells in vivo, the HA-HP hydrogel with AFS cells improved wound closure and reepithelialization and increased vascularization and production of extracellular matrix in vivo. These results suggest that HA-HP hydrogel has the potential to prolong the paracrine activity of cells, thereby increasing their therapeutic effectiveness in wound healing. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 105B: 1986-2000, 2017.
Collapse
Affiliation(s)
- Aleksander Skardal
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina.,Department of Biomedical Engineering, Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, Wake Forest Baptist Health, Medical Center Boulevard, Winston-Salem, North Carolina.,Department of Cancer Biology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina
| | - Sean V Murphy
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina
| | - Kathryn Crowell
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina
| | - David Mack
- Department of Rehabilitation Medicine, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington.,Department of Bioengineering, University of Washington, Seattle, Washington
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina.,Department of Biomedical Engineering, Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, Wake Forest Baptist Health, Medical Center Boulevard, Winston-Salem, North Carolina
| | - Shay Soker
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina.,Department of Biomedical Engineering, Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, Wake Forest Baptist Health, Medical Center Boulevard, Winston-Salem, North Carolina.,Department of Cancer Biology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina
| |
Collapse
|
657
|
Darkazalli A, Vied C, Badger CD, Levenson CW. Human Mesenchymal Stem Cell Treatment Normalizes Cortical Gene Expression after Traumatic Brain Injury. J Neurotrauma 2016; 34:204-212. [PMID: 27161121 DOI: 10.1089/neu.2015.4322] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Traumatic brain injury (TBI) results in a progressive disease state with many adverse and long-term neurological consequences. Mesenchymal stem cells (MSCs) have emerged as a promising cytotherapy and have been previously shown to reduce secondary apoptosis and cognitive deficits associated with TBI. Consistent with the established literature, we observed that systemically administered human MSCs (hMSCs) accumulate with high specificity at the TBI lesion boundary zone known as the penumbra. Substantial work has been done to illuminate the mechanisms by which MSCs, and the bioactive molecules they secrete, exert their therapeutic effect. However, no such work has been published to examine the effect of MSC treatment on gene expression in the brain post-TBI. In the present study, we use high-throughput RNA sequencing (RNAseq) of cortical tissue from the TBI penumbra to assess the molecular effects of both TBI and subsequent treatment with intravenously delivered hMSCs. RNAseq revealed that expression of almost 7000 cortical genes in the penumbra were differentially regulated by TBI. Pathway analysis using the KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway database revealed that TBI regulated a large number of genes belonging to pathways involved in metabolism, receptor-mediated cell signaling, neuronal plasticity, immune cell recruitment and infiltration, and neurodegenerative disease. Remarkably, hMSC treatment was found to normalize 49% of all genes disrupted by TBI, with notably robust normalization of specific pathways within the categories mentioned above, including neuroactive receptor-ligand interactions (57%), glycolysis and gluconeogenesis (81%), and Parkinson's disease (100%). These data provide evidence in support of the multi-mechanistic nature of stem cell therapy and suggest that hMSC treatment is capable of simultaneously normalizing a wide variety of important molecular pathways that are disrupted by brain injury.
Collapse
Affiliation(s)
- Ali Darkazalli
- 1 Department of Biomedical Sciences, Florida State University College of Medicine , Tallahassee, Florida.,2 Program in Neuroscience, Florida State University College of Medicine , Tallahassee, Florida
| | - Cynthia Vied
- 1 Department of Biomedical Sciences, Florida State University College of Medicine , Tallahassee, Florida
| | - Crystal-Dawn Badger
- 1 Department of Biomedical Sciences, Florida State University College of Medicine , Tallahassee, Florida
| | - Cathy W Levenson
- 1 Department of Biomedical Sciences, Florida State University College of Medicine , Tallahassee, Florida.,2 Program in Neuroscience, Florida State University College of Medicine , Tallahassee, Florida
| |
Collapse
|
658
|
Abstract
Temporomandibular Disorders (TMD) represent a heterogeneous group of musculoskeletal and neuromuscular conditions involving the temporomandibular joint (TMJ), masticatory muscles and/or associated structures. They are a major cause of non-dental orofacial pain. As a group, they are often multi-factorial in nature and have no common etiology or biological explanations. TMD can be broadly divided into masticatory muscle and TMJ disorders. TMJ disorders are characterized by intra-articular positional and/or structural abnormalities. The most common type of TMJ disorders involves displacement of the TMJ articular disc that precedes progressive degenerative changes of the joint leading to osteoarthritis (OA). In the past decade, progress made in the development of stem cell-based therapies and tissue engineering have provided alternative methods to attenuate the disease symptoms and even replace the diseased tissue in the treatment of TMJ disorders. Resident mesenchymal stem cells (MSCs) have been isolated from the synovia of TMJ, suggesting an important role in the repair and regeneration of TMJ. The seminal discovery of pluripotent stem cells including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) have provided promising cell sources for drug discovery, transplantation as well as for tissue engineering of TMJ condylar cartilage and disc. This review discusses the most recent advances in development of stem cell-based treatments for TMJ disorders through innovative approaches of cell-based therapeutics, tissue engineering and drug discovery.
Collapse
|
659
|
3D-Printed PCL Scaffolds for the Cultivation of Mesenchymal Stem Cells. J Appl Biomater Funct Mater 2016; 14:e19-25. [DOI: 10.5301/jabfm.5000252] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2015] [Indexed: 11/20/2022] Open
Abstract
Introduction Tissue engineering is a field which is currently under a great deal of investigation for the development and/or restoration of tissue and organs, through the combination of cell therapy with biomaterials. Rapid prototyping or additive manufacturing is a versatile technology which makes possible the fabrication of three dimensional (3D) structures from a wide range of materials with complex geometry and accuracy, such as scaffolds. Aim The aim of this study has been to investigate the interaction between mesenchymal stem cells with poly (ε-caprolactone) (PCL) biomaterials used for obtaining scaffolds through additive manufacturing. Materials and Methods Scanning electron microscopy, confocal microscopy and biological assays were performed to analyse the successful interaction between the cells and the biomaterials. Results As a result, the number of viable cells attached to the scaffolds was lower when compared to the control group; however, it was possible to observe cells in the scaffolds since day 1 of analysis, with regions of confluence after 21 days of seeding. Conclusions To conclude, these biomaterials are interesting if used as medical artifacts, principally in tissue with prolonged regeneration time and which requires 3D supports with good mechanical properties.
Collapse
|
660
|
Luciani N, Du V, Gazeau F, Richert A, Letourneur D, Le Visage C, Wilhelm C. Successful chondrogenesis within scaffolds, using magnetic stem cell confinement and bioreactor maturation. Acta Biomater 2016; 37:101-10. [PMID: 27063490 DOI: 10.1016/j.actbio.2016.04.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 04/05/2016] [Accepted: 04/06/2016] [Indexed: 01/24/2023]
Abstract
UNLABELLED Tissue engineering strategies, such as cellularized scaffolds approaches, have been explored for cartilage replacement. The challenge, however, remains to produce a cartilaginous tissue incorporating functional chondrocytes and being large and thick enough to be compatible with the replacement of articular defects. Here, we achieved unprecedented cartilage tissue production into a porous polysaccharide scaffold by combining of efficient magnetic condensation of mesenchymal stem cells, and dynamic maturation in a bioreactor. In optimal conditions, all the hallmarks of chondrogenesis were enhanced with a 50-fold increase in collagen II expression compared to negative control, an overexpression of aggrecan and collagen XI, and a very low expression of collagen I and RUNX2. Histological staining showed a large number of cellular aggregates, as well as an increased proteoglycan synthesis by chondrocytes. Interestingly, electron microscopy showed larger chondrocytes and a more abundant extracellular matrix. In addition, the periodicity of the neosynthesized collagen fibers matched that of collagen II. These results represent a major step forward in replacement tissue for cartilage defects. STATEMENT OF SIGNIFICANCE A combination of several innovative technologies (magnetic cell seeding, polysaccharide porous scaffolds, and dynamic maturation in bioreactor) enabled unprecedented successful chondrogenesis within scaffolds.
Collapse
Affiliation(s)
- Nathalie Luciani
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057 CNRS & University Paris Diderot, Paris F-75205 Cedex 13, France.
| | - Vicard Du
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057 CNRS & University Paris Diderot, Paris F-75205 Cedex 13, France
| | - Florence Gazeau
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057 CNRS & University Paris Diderot, Paris F-75205 Cedex 13, France
| | - Alain Richert
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057 CNRS & University Paris Diderot, Paris F-75205 Cedex 13, France
| | - Didier Letourneur
- Laboratoire de recherche vasculaire translationnelle, INSERM UMR 1148 & University Paris Diderot, Paris, France
| | | | - Claire Wilhelm
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057 CNRS & University Paris Diderot, Paris F-75205 Cedex 13, France
| |
Collapse
|
661
|
Badner A, Vawda R, Laliberte A, Hong J, Mikhail M, Jose A, Dragas R, Fehlings M. Early Intravenous Delivery of Human Brain Stromal Cells Modulates Systemic Inflammation and Leads to Vasoprotection in Traumatic Spinal Cord Injury. Stem Cells Transl Med 2016; 5:991-1003. [PMID: 27245367 DOI: 10.5966/sctm.2015-0295] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 03/07/2016] [Indexed: 12/19/2022] Open
Abstract
UNLABELLED : Spinal cord injury (SCI) is a life-threatening condition with multifaceted complications and limited treatment options. In SCI, the initial physical trauma is closely followed by a series of secondary events, including inflammation and blood spinal cord barrier (BSCB) disruption, which further exacerbate injury. This secondary pathology is partially mediated by the systemic immune response to trauma, in which cytokine production leads to the recruitment/activation of inflammatory cells. Because early intravenous delivery of mesenchymal stromal cells (MSCs) has been shown to mitigate inflammation in various models of neurologic disease, this study aimed to assess these effects in a rat model of SCI (C7-T1, 35-gram clip compression) using human brain-derived stromal cells. Quantitative polymerase chain reaction for a human-specific DNA sequence was used to assess cell biodistribution/clearance and confirmed that only a small proportion (approximately 0.001%-0.002%) of cells are delivered to the spinal cord, with the majority residing in the lung, liver, and spleen. Intriguingly, although cell populations drastically declined in all aforementioned organs, there remained a persistent population in the spleen at 7 days. Furthermore, the cell infusion significantly increased splenic and circulating levels of interleukin-10-a potent anti-inflammatory cytokine. Through this suppression of the systemic inflammatory response, the cells also reduced acute spinal cord BSCB permeability, hemorrhage, and lesion volume. These early effects further translated into enhanced functional recovery and tissue sparing 10 weeks after SCI. This work demonstrates an exciting therapeutic approach whereby a minimally invasive cell-transplantation procedure can effectively reduce secondary damage after SCI through systemic immunomodulation. SIGNIFICANCE Central nervous system pericytes (perivascular stromal cells) have recently gained significant attention within the scientific community. In addition to being recognized as major players in neurotrauma, pericytes have been discovered to share a common origin and potentially function with traditionally defined mesenchymal stromal cells (MSCs). Although there have been several in vitro comparisons, the in vivo therapeutic application of human brain-derived stromal cells has not been previously evaluated. This study demonstrates that these cells not only display a MSC phenotype in vitro but also have similar in vivo immunomodulatory effects after spinal cord injury that are more potent than those of non-central nervous system tissue-derived cells. Therefore, these cells are of great interest for therapeutic use in spinal cord injury.
Collapse
Affiliation(s)
- Anna Badner
- Division of Genetics and Development, Toronto Western Research Institute, University Health Network, Toronto, Toronto, Ontario, Canada Institute of Medical Science, University of Toronto, Ontario, Canada
| | - Reaz Vawda
- Division of Genetics and Development, Toronto Western Research Institute, University Health Network, Toronto, Toronto, Ontario, Canada
| | - Alex Laliberte
- Division of Genetics and Development, Toronto Western Research Institute, University Health Network, Toronto, Toronto, Ontario, Canada Institute of Medical Science, University of Toronto, Ontario, Canada
| | - James Hong
- Division of Genetics and Development, Toronto Western Research Institute, University Health Network, Toronto, Toronto, Ontario, Canada Institute of Medical Science, University of Toronto, Ontario, Canada
| | - Mirriam Mikhail
- Division of Genetics and Development, Toronto Western Research Institute, University Health Network, Toronto, Toronto, Ontario, Canada
| | - Alejandro Jose
- Division of Genetics and Development, Toronto Western Research Institute, University Health Network, Toronto, Toronto, Ontario, Canada
| | - Rachel Dragas
- Division of Genetics and Development, Toronto Western Research Institute, University Health Network, Toronto, Toronto, Ontario, Canada Institute of Medical Science, University of Toronto, Ontario, Canada
| | - Michael Fehlings
- Division of Genetics and Development, Toronto Western Research Institute, University Health Network, Toronto, Toronto, Ontario, Canada Institute of Medical Science, University of Toronto, Ontario, Canada Spinal Program, University Health Network, Toronto Western Hospital, Toronto, Ontario, Canada
| |
Collapse
|
662
|
Sweeney NP, Regan C, Liu J, Galleu A, Dazzi F, Lindemann D, Rupar CA, McClure MO. Rapid and Efficient Stable Gene Transfer to Mesenchymal Stromal Cells Using a Modified Foamy Virus Vector. Mol Ther 2016; 24:1227-36. [PMID: 27133965 PMCID: PMC4982542 DOI: 10.1038/mt.2016.91] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 04/19/2016] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) hold great promise for regenerative medicine. Stable ex vivo gene transfer to MSCs could improve the outcome and scope of MSC therapy, but current vectors require multiple rounds of transduction, involve genotoxic viral promoters and/or the addition of cytotoxic cationic polymers in order to achieve efficient transduction. We describe a self-inactivating foamy virus vector (FVV), incorporating the simian macaque foamy virus envelope and using physiological promoters, which efficiently transduces murine MSCs (mMSCs) in a single-round. High and sustained expression of the transgene, whether GFP or the lysosomal enzyme, arylsulphatase A (ARSA), was achieved. Defining MSC characteristics (surface marker expression and differentiation potential), as well as long-term engraftment and distribution in the murine brain following intracerebroventricular delivery, are unaffected by FVV transduction. Similarly, greater than 95% of human MSCs (hMSCs) were stably transduced using the same vector, facilitating human application. This work describes the best stable gene transfer vector available for mMSCs and hMSCs.
Collapse
Affiliation(s)
- Nathan Paul Sweeney
- Jefferiss Research Trust laboratories, Department of Medicine, Imperial College London, London, UK
| | - Cathy Regan
- Department of Pathology and Laboratory Medicine, Western University, Ontario, Canada.,Department of Biochemistry and Pediatrics, Western University, Ontario, Canada
| | - Jiahui Liu
- Department of Pathology and Laboratory Medicine, Western University, Ontario, Canada.,Department of Biochemistry and Pediatrics, Western University, Ontario, Canada
| | - Antonio Galleu
- Department of Haemato-Oncology, King's College London, London, UK
| | - Francesco Dazzi
- Department of Haemato-Oncology, King's College London, London, UK
| | - Dirk Lindemann
- Institute of Virology, Technische Universität Dresden, Dresden, Germany
| | - Charles Anthony Rupar
- Department of Pathology and Laboratory Medicine, Western University, Ontario, Canada.,Department of Biochemistry and Pediatrics, Western University, Ontario, Canada
| | - Myra Olga McClure
- Jefferiss Research Trust laboratories, Department of Medicine, Imperial College London, London, UK
| |
Collapse
|
663
|
Pirzad Jahromi G, Shabanzadeh Pirsaraei A, Sadr SS, Kaka G, Jafari M, Seidi S, Charish J. Multipotent bone marrow stromal cell therapy promotes endogenous cell proliferation following ischemic stroke. Clin Exp Pharmacol Physiol 2016. [PMID: 26218989 DOI: 10.1111/1440-1681.12466] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Despite extensive research over the years, there still exists some debate as to what constitutes the optimal therapeutic strategy to promote recovery following stroke. Due to the complexity of injured brain pathophysiology, treatment approaches should ideally address numerous factors, ultimately aiming to promote tissue protection, axonal regrowth and functional recovery. This study extends the understanding of the effects of bone marrow stromal cell (BMSC) treatment following experimentally induced ischemic stroke in rats. Focal ischemic brain injury was experimentally induced in rats by placing a preformed clot into the middle cerebral artery. Animals were injected intravenously with BMSCs at 24 h after stroke and were killed 7 days post injury. When administered BMSCs following stroke, the neurological outcome was significantly improved relative to controls. There was an increase in the number of BMSCs labelled with BrdU present in the injured hemisphere of the brain compared to the non-injured side. Furthermore, administration of BMSCs also led to increases in astrocytosis, vascularization and endogenous proliferation. These findings provide insight into the mechanisms of action of BMSC treatment and further argue for the therapeutic potential of BMSCs as an effective treatment following cerebral stroke.
Collapse
Affiliation(s)
- Gila Pirzad Jahromi
- Neuroscience Research Centre, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Alireza Shabanzadeh Pirsaraei
- Electrophysiology Research Centre, Neuroscience Institute, Tehran, Iran.,Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Genetics and Development Division, Toronto Western Research Institute, Toronto, ON, Canada
| | - Seyed Shahabeddin Sadr
- Electrophysiology Research Centre, Neuroscience Institute, Tehran, Iran.,Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Golamreza Kaka
- Neuroscience Research Centre, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mahvash Jafari
- Department of Biochemistry, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Sadegh Seidi
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Jason Charish
- Genetics and Development Division, Toronto Western Research Institute, Toronto, ON, Canada
| |
Collapse
|
664
|
Archibald PR, Chandra A, Thomas D, Morley G, Lekishvili T, Devonshire A, Williams DJ. Comparability of scalable, automated hMSC culture using manual and automated process steps. Biochem Eng J 2016. [DOI: 10.1016/j.bej.2015.07.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
665
|
Samsonraj RM, Rai B, Sathiyanathan P, Puan KJ, Rötzschke O, Hui JH, Raghunath M, Stanton LW, Nurcombe V, Cool SM. Establishing criteria for human mesenchymal stem cell potency. Stem Cells 2016; 33:1878-91. [PMID: 25752682 PMCID: PMC5363381 DOI: 10.1002/stem.1982] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 01/08/2015] [Indexed: 12/15/2022]
Abstract
This study sought to identify critical determinants of mesenchymal stem cell (MSC) potency using in vitro and in vivo attributes of cells isolated from the bone marrow of age‐ and sex‐matched donors. Adherence to plastic was not indicative of potency, yet capacity for long‐term expansion in vitro varied considerably between donors, allowing the grouping of MSCs from the donors into either those with high‐growth capacity or low‐growth capacity. Using this grouping strategy, high‐growth capacity MSCs were smaller in size, had greater colony‐forming efficiency, and had longer telomeres. Cell‐surface biomarker analysis revealed that the International Society for Cellular Therapy (ISCT) criteria did not distinguish between high‐growth capacity and low‐growth capacity MSCs, whereas STRO‐1 and platelet‐derived growth factor receptor alpha were preferentially expressed on high‐growth capacity MSCs. These cells also had the highest mean expression of the mRNA transcripts TWIST‐1 and DERMO‐1. Irrespective of these differences, both groups of donor MSCs produced similar levels of key growth factors and cytokines involved in tissue regeneration and were capable of multilineage differentiation. However, high‐growth capacity MSCs produced approximately double the volume of mineralized tissue compared to low‐growth capacity MSCs when assessed for ectopic bone‐forming ability. The additional phenotypic criteria presented in this study when combined with the existing ISCT minimum criteria and working proposal will permit an improved assessment of MSC potency and provide a basis for establishing the quality of MSCs prior to their therapeutic application. Stem Cells2015;33:1878–1891
Collapse
Affiliation(s)
| | - Bina Rai
- Glycotherapeutics Group.,Sciences, Singapore University of Technology and Design, 8 Somapah Road, Singapore
| | - Padmapriya Sathiyanathan
- Stem Cell and Regenerative Biology, Genome Institute of Singapore, A*STAR, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore
| | - Kia Joo Puan
- Singapore Immunology Network (SIgN), A*STAR, Singapore
| | | | - James H Hui
- Department of Orthopedic Surgery, National University of Singapore, Singapore
| | - Michael Raghunath
- Advanced Wound Care Laboratory, Institute of Medical Biology, A*STAR, Singapore.,Department of Biomedical Engineering.,Department of Biochemistry.,NUS Tissue Engineering Programme
| | - Lawrence W Stanton
- Department of Biological Sciences, National University of Singapore, Singapore.,Stem Cell and Regenerative Biology, Genome Institute of Singapore, A*STAR, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore
| | - Victor Nurcombe
- Glycotherapeutics Group.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Simon M Cool
- Glycotherapeutics Group.,Department of Orthopedic Surgery, National University of Singapore, Singapore
| |
Collapse
|
666
|
Abstract
Silicosis is the most common pneumoconiosis globally, with higher prevalence and incidence in developing countries. To date, there is no effective treatment to halt or reverse the disease progression caused by silica-induced lung injury. Significant advances have to be made in order to reduce morbidity and mortality related to silicosis. In this review, we have highlighted the main mechanisms of action that cause lung damage by silica particles and summarized the data concerning the therapeutic promise of cell-based therapy for silicosis.
Collapse
|
667
|
Hirose Y, Yamamoto T, Nakashima M, Funahashi Y, Matsukawa Y, Yamaguchi M, Kawabata S, Gotoh M. Injection of Dental Pulp Stem Cells Promotes Healing of Damaged Bladder Tissue in a Rat Model of Chemically Induced Cystitis. Cell Transplant 2016; 25:425-36. [DOI: 10.3727/096368915x689523] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Dental pulp stem cells (DPSCs) are reported as sources of mesenchymal stem cells (MSCs). MSCs are used as cell therapy options for various diseases. The present study examined the healing effects of DPSC injection on damaged bladder tissue in a chemically induced cystitis rat model. Cystitis was induced by hydrochloride injection into the bladder of female F344/NSlc rats. On the following day, DPSCs suspended in phosphate-buffered saline (PBS) were injected into the bladder and maintained for 1 h (DPSC injection group), while PBS alone was injected as the standard for comparison (PBS injection group). After 2 days following injection, considerable submucosal edema, vascular structure destruction, hemorrhage, and inflammatory cell invasion were observed both in the DPSC and PBS injection groups, with no difference in their degree of submucosal edema and hemorrhage. Six days after injection, vascular structure regeneration was observed in both groups; however, unlike the DPSC injection group, the PBS injection group showed traces of submucosal edema and hemorrhage. These results correlated with tissue concentrations of myeloperoxidase (MPO) and the inflammatory cytokines IL-1β, IL-6, and TNF-α. Furthermore, the intercontraction interval was prolonged, and the frequency of nociceptive behaviors was reduced in the DPSC injection group compared with the PBS injection group. DPSCs were found on the bladder epithelium until day 3 after injection. In the DPSC-conditioned media (CM), the trophic factors FGF-2, VEGF, and the C-C and C-X-C families of chemokines were detected. The results of DPSC injection into the cystitis rat model suggested that the injected cells promote the healing of the damaged bladder tissue by exerting various trophic effects while localizing on the bladder epithelium and that MSC injection is a potential novel therapy for interstitial cystitis/painful bladder syndrome.
Collapse
Affiliation(s)
- Yujiro Hirose
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Dental Regenerative Medicine, Center of Advanced Medicine for Dental and Oral Diseases, National Center for Geriatrics and Gerontology, Obu, Japan
- Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry, Suita, Japan
| | - Tokunori Yamamoto
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Misako Nakashima
- Department of Dental Regenerative Medicine, Center of Advanced Medicine for Dental and Oral Diseases, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Yasuhito Funahashi
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshihisa Matsukawa
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masaya Yamaguchi
- Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry, Suita, Japan
| | - Shigetada Kawabata
- Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry, Suita, Japan
| | - Momokazu Gotoh
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
668
|
Barrachina L, Remacha A, Romero A, Vázquez F, Albareda J, Prades M, Ranera B, Zaragoza P, Martín-Burriel I, Rodellar C. Effect of inflammatory environment on equine bone marrow derived mesenchymal stem cells immunogenicity and immunomodulatory properties. Vet Immunol Immunopathol 2016; 171:57-65. [DOI: 10.1016/j.vetimm.2016.02.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 02/03/2016] [Accepted: 02/07/2016] [Indexed: 02/07/2023]
|
669
|
Najar M, Raicevic G, André T, Fayyad-Kazan H, Pieters K, Bron D, Toungouz M, Lagneaux L. Mesenchymal stromal cells from the foreskin: Tissue isolation, cell characterization and immunobiological properties. Cytotherapy 2016; 18:320-335. [PMID: 26857227 DOI: 10.1016/j.jcyt.2015.11.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Revised: 11/12/2015] [Accepted: 11/16/2015] [Indexed: 12/21/2022]
Abstract
BACKGROUND AIMS Because of their self-renewal capacity, multilineage potential and immunomodulatory properties, MSCs are an attractive tool for cell-based immunotherapy strategies. Foreskin, considered as a biological waste material, has been shown to be a reservoir of therapeutic cells. METHODS MSCs were isolated from different foreskin samples, maintained under in vitro culture and defined according to the International Society for Cellular Therapy (ISCT) criteria. We subsequently determined their main cell characteristics as well as their immunobiological properties. The following parameters were determined: (i) morphology and phenotype, (ii) proliferative and clonogenic potentials, (iii) tri-lineage differentiation ability, (iv) immunological profile, (v) immunomodulatory properties and (vi) protein and messenger RNA expression/secretion profile of immunoregulatory cytokines/factors as well as the pattern of toll-like receptors (TLRs). By using a pro-inflammatory cytokine cocktail, we also evaluated the influence of an inflammatory environment on their biology. RESULTS With a typical fibroblast-like morphology and an ISCT-compliant phenotype, foreskin-MSCs (FSK-MSCs) were highly proliferative and had a great clonogenic potential. They displayed multilineage capacities and interesting immunomodulatory properties. Of importance, FSK-MSCs were not immunogenetic and were further able to inhibit T-cell proliferation. We showed that several immunoregulatory cytokines and factors might be potentially involved in FSK-MSC immunomodulation with particular attention to hepatocyte growth factor and interleukin-11. Moreover, FSK-MSCs expressed several TLRs and were sensitive to the inflammatory environment by properly adjusting their profile and fate. CONCLUSIONS Foreskin represents a new alternative source for MSCs that is compliant with ISCT criteria. Their unique immunobiological properties allow consideration of FSK-MSCs as a valuable tolerogenic product for cell-based immunotherapy.
Collapse
Affiliation(s)
- Mehdi Najar
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles, Campus Erasme, Brussels, Belgium.
| | - Gordana Raicevic
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles, Campus Erasme, Brussels, Belgium
| | - Thibaud André
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles, Campus Erasme, Brussels, Belgium
| | - Hussein Fayyad-Kazan
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles, Campus Erasme, Brussels, Belgium
| | - Karlien Pieters
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles, Campus Erasme, Brussels, Belgium
| | - Dominique Bron
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles, Campus Erasme, Brussels, Belgium
| | - Michel Toungouz
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles, Campus Erasme, Brussels, Belgium
| | - Laurence Lagneaux
- Laboratory of Clinical Cell Therapy, Institut Jules Bordet, Université Libre de Bruxelles, Campus Erasme, Brussels, Belgium
| |
Collapse
|
670
|
Abumaree MH, Abomaray FM, Alshehri NA, Almutairi A, AlAskar AS, Kalionis B, Al Jumah MA. Phenotypic and Functional Characterization of Mesenchymal Stem/Multipotent Stromal Cells From Decidua Parietalis of Human Term Placenta. Reprod Sci 2016; 23:1193-207. [PMID: 26902429 DOI: 10.1177/1933719116632924] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Mesenchymal stem/multipotent stromal cells (MSCs) from the human placenta show stem cell-like properties useful for regenerative medicine. Previously, we reported that MSCs isolated from the fetal part of human term placentae have characteristics, which make them a potential candidate for regenerative medicine. In this study, we characterized MSC isolated from the maternal part of human term placenta. The MSCs were isolated from the decidua parietalis (DPMSCs) of human placenta using a digestion method and characterized by colony-forming unit assay and the expression of MSC markers by flow cytometry technique. In addition, DPMSC differentiation into the 3 mesenchymal lineages was also performed. Moreover, the gene and protein expression profiles of DPMSCs were identified by real-time polymerase chain reaction and flow cytometry techniques, respectively. Furthermore, proteins secreted by DPMSCs were detected by sandwich enzyme-linked immunosorbent assays. Finally, the proliferation and migration potentials of DPMSCs were also determined. The DPMSCs were positive for MSC markers and negative for hematopoietic and endothelial markers, as well as costimulatory molecules and HLA-DR. Functionally, DPMSCs formed colonies and differentiated into chondrocytes, osteocytes, and adipocytes. In addition, they proliferated and migrated in response to different stimuli. Finally, they expressed and secreted many biological and immunological factors with multiple functions. Here, we carry out an extensive characterization of DPMSCs of human placenta. We report that these cells express and secrete a wide range of molecules with multiple functions, and therefore, we suggest that these cells could be an attractive candidate for cell-based therapy.
Collapse
Affiliation(s)
- Mohamed H Abumaree
- College of Science and Health Professions, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - F M Abomaray
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - N A Alshehri
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - A Almutairi
- College of Applied Medical Sciences, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - A S AlAskar
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - B Kalionis
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, Australia Department of Perinatal Medicine Pregnancy Research Centre, Royal Women's Hospital, Parkville, Victoria, Australia
| | - M A Al Jumah
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| |
Collapse
|
671
|
Lavandula angustifolia Extract Improves the Result of Human Umbilical Mesenchymal Wharton's Jelly Stem Cell Transplantation after Contusive Spinal Cord Injury in Wistar Rats. Stem Cells Int 2016; 2016:5328689. [PMID: 27057171 PMCID: PMC4769777 DOI: 10.1155/2016/5328689] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 01/10/2016] [Indexed: 11/17/2022] Open
Abstract
Introduction. The primary trauma of spinal cord injury (SCI) results in severe damage to nervous functions. At the cellular level, SCI causes astrogliosis. Human umbilical mesenchymal stem cells (HUMSCs), isolated from Wharton's jelly of the umbilical cord, can be easily obtained. Previously, we showed that the neuroprotective effects of Lavandula angustifolia can lead to improvement in a contusive SCI model in rats. Objective. The aim of this study was to investigate the effect of L. angustifolia (Lav) on HUMSC transplantation after acute SCI. Materials and Methods. Sixty adult female rats were randomly divided into eight groups. Every week after SCI onset, all animals were evaluated for behavior outcomes. H&E staining was performed to examine the lesions after injury. GFAP expression was assessed for astrogliosis. Somatosensory evoked potential (SEP) testing was performed to detect the recovery of neural conduction. Results. Behavioral tests showed that the HUMSC group improved in comparison with the SCI group, but HUMSC + Lav 400 was very effective, resulting in a significant increase in locomotion activity. Sensory tests and histomorphological and immunohistochemistry analyses verified the potentiation effects of Lav extract on HUMSC treatment. Conclusion. Transplantation of HUMSCs is beneficial for SCI in rats, and Lav extract can potentiate the functional and cellular recovery with HUMSC treatment in rats after SCI.
Collapse
|
672
|
Phenotypic and Functional Characterization of Mesenchymal Stem/Multipotent Stromal Cells from Decidua Basalis of Human Term Placenta. Stem Cells Int 2016; 2016:5184601. [PMID: 27087815 PMCID: PMC4764756 DOI: 10.1155/2016/5184601] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 12/14/2015] [Accepted: 01/05/2016] [Indexed: 01/10/2023] Open
Abstract
Mesenchymal stem cell (MSC) therapies for the treatment of diseases associated with inflammation and oxidative stress employ primarily bone marrow MSCs (BMMSCs) and other MSC types such as MSC from the chorionic villi of human term placentae (pMSCs). These MSCs are not derived from microenvironments associated with inflammation and oxidative stress, unlike MSCs from the decidua basalis of the human term placenta (DBMSCs). DBMSCs were isolated and then extensively characterized. Differentiation of DBMSCs into three mesenchymal lineages (adipocytes, osteocytes, and chondrocytes) was performed. Real-time polymerase chain reaction (PCR) and flow cytometry techniques were also used to characterize the gene and protein expression profiles of DBMSCs, respectively. In addition, sandwich enzyme-linked immunosorbent assay (ELISA) was performed to detect proteins secreted by DBMSCs. Finally, the migration and proliferation abilities of DBMSCs were also determined. DBMSCs were positive for MSC markers and HLA-ABC. DBMSCs were negative for hematopoietic and endothelial markers, costimulatory molecules, and HLA-DR. Functionally, DBMSCs differentiated into three mesenchymal lineages, proliferated, and migrated in response to a number of stimuli. Most importantly, these cells express and secrete a distinct combination of cytokines, growth factors, and immune molecules that reflect their unique microenvironment. Therefore, DBMSCs could be attractive, alternative candidates for MSC-based therapies that treat diseases associated with inflammation and oxidative stress.
Collapse
|
673
|
Merino-González C, Zuñiga FA, Escudero C, Ormazabal V, Reyes C, Nova-Lamperti E, Salomón C, Aguayo C. Mesenchymal Stem Cell-Derived Extracellular Vesicles Promote Angiogenesis: Potencial Clinical Application. Front Physiol 2016; 7:24. [PMID: 26903875 PMCID: PMC4746282 DOI: 10.3389/fphys.2016.00024] [Citation(s) in RCA: 167] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 01/18/2016] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are adult multipotent stem cells that are able to differentiate into multiple specialized cell types including osteocytes, adipocytes, and chondrocytes. MSCs exert different functions in the body and have recently been predicted to have a major clinical/therapeutic potential. However, the mechanisms of self-renewal and tissue regeneration are not completely understood. It has been shown that the biological effect depends mainly on its paracrine action. Furthermore, it has been reported that the secretion of soluble factors and the release of extracellular vesicles, such as exosomes, could mediate the cellular communication to induce cell-differentiation/self-renewal. This review provides an overview of MSC-derived exosomes in promoting angiogenicity and of the clinical relevance in a therapeutic approach.
Collapse
Affiliation(s)
- Consuelo Merino-González
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepción Concepción, Chile
| | - Felipe A Zuñiga
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepción Concepción, Chile
| | - Carlos Escudero
- Vascular Physiology Laboratory, Group of Investigation in Tumor Angiogenesis (GIANT), Department of Basic Sciences, Universidad del Bío-BíoChillán, Chile; Group of Research and Innovation in Vascular Health (GRIVAS Health)Chillán, Chile
| | - Valeska Ormazabal
- Department of Physiopathology, Faculty of Biological Sciences, University of Concepción Concepción, Chile
| | - Camila Reyes
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of Concepción Concepción, Chile
| | | | - Carlos Salomón
- Exosome Biology Laboratory, Centre for Clinical Diagnostics, University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital, The University of Queensland Brisbane, QLD, Australia
| | - Claudio Aguayo
- Department of Clinical Biochemistry and Immunology, Faculty of Pharmacy, University of ConcepciónConcepción, Chile; Group of Research and Innovation in Vascular Health (GRIVAS Health)Chillán, Chile
| |
Collapse
|
674
|
Huang B, Cheng X, Wang H, Huang W, la Ga Hu Z, Wang D, Zhang K, Zhang H, Xue Z, Da Y, Zhang N, Hu Y, Yao Z, Qiao L, Gao F, Zhang R. Mesenchymal stem cells and their secreted molecules predominantly ameliorate fulminant hepatic failure and chronic liver fibrosis in mice respectively. J Transl Med 2016; 14:45. [PMID: 26861623 PMCID: PMC4746907 DOI: 10.1186/s12967-016-0792-1] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 01/20/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Orthotopic liver transplantation is the only effective treatment for liver failure but limited with shortage of available donor organs. Recent studies show promising results of mesenchymal stem cells (MSCs)-based therapies. METHODS We systematically investigate the therapeutic effects of MSCs or MSC-conditioned medium (MSC-CM) in ameliorating fulminant hepatic failure (FHF) and chronic liver fibrosis in mice. In addition, extensive flow cytometry analysis of spleens from vehicle and MSC- and MSC-CM-treated mice was applied to reveal the alteration of inflammatory state. RESULTS In FHF model, MSCs treatment reduced remarkably the death incidents; the analysis of gross histopathology showed that control livers were soft and shrunken with extensive extravasated blood, which was gradually reduced at later time points, while MSC-treated livers showed gross pathological changes, even 24 h after MSC infusion, and hematoxylin and eosin staining revealed dramatical hepatocellular death with cytoplasmic vacuolization suppressed by MSCs treatment; flow cytometry analysis of total lymphocytes showed that macrophages (F4/80) infiltrated into control livers more than MSC-treated livers; by contrast, MSC-CM partially ameliorates FHF. In chronic liver injury model, MSC and MSC-CM both suppressed fibrogenesis and necroinflammatory, and the later was better; activation of hepatic stellate cells (α-SMA) was inhibited; glycogen synthesis and storage (indicated by periodic acid-Schiff -staining) was improved; liver regeneration (Ki67) was promoted while liver apoptosis (TUNEL) was reduced. In the in vitro, MSCs promote macrophage line RAW264.7 apoptosis and MSC-CM promotes apoptosis and inhibits proliferation of HSC line LX-2. We also found that MSCs and MSC-CM could improve spleen; MSC-CM increased levels of Th2 and Treg cells, and reduced levels of Th17 cells, whereas levels of Th1 cells were unchanged; comparatively, MSC treatment did not affect Th17 and Treg cells and only slightly alters inflammatory state; MSC and MSC-CM treatment both substantially down-regulated macrophages in the spleens. CONCLUSION Both MSCs and MSC-CM exert therapeutic effects by acting on various key cells during the pathogenesis of FHF and chronic fibrosis, stimulating hepatocyte proliferation and suppressing apoptosis, down-regulating infiltrating macrophages, converting CD4(+) T lymphocyte system into an anti-inflammatory state, and facilitating hepatic stellate cell death.
Collapse
Affiliation(s)
- Biao Huang
- Department of Immunology and Research Center of Basic Medical Science; Tianjin Key Laboratory of Cellular and Molecular Immunology; Key Laboratory of Immune Microenvironment and Diseases, Ministry of Education of China, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Qi Xiang Tai Road No.22, Tianjin, 300070, China.
| | - Xixi Cheng
- Department of Immunology and Research Center of Basic Medical Science; Tianjin Key Laboratory of Cellular and Molecular Immunology; Key Laboratory of Immune Microenvironment and Diseases, Ministry of Education of China, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Qi Xiang Tai Road No.22, Tianjin, 300070, China.
| | - Huafeng Wang
- Department of Immunology and Research Center of Basic Medical Science; Tianjin Key Laboratory of Cellular and Molecular Immunology; Key Laboratory of Immune Microenvironment and Diseases, Ministry of Education of China, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Qi Xiang Tai Road No.22, Tianjin, 300070, China.
- School of Life Science, Shanxi Normal University, Linfen, Shanxi Province, China.
| | - Wenjing Huang
- Department of Immunology and Research Center of Basic Medical Science; Tianjin Key Laboratory of Cellular and Molecular Immunology; Key Laboratory of Immune Microenvironment and Diseases, Ministry of Education of China, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Qi Xiang Tai Road No.22, Tianjin, 300070, China.
| | - Zha la Ga Hu
- Department of Cell Biology, Logistic College of CAPF, Tianjin, China.
| | - Dan Wang
- Department of Immunology and Research Center of Basic Medical Science; Tianjin Key Laboratory of Cellular and Molecular Immunology; Key Laboratory of Immune Microenvironment and Diseases, Ministry of Education of China, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Qi Xiang Tai Road No.22, Tianjin, 300070, China.
| | - Kai Zhang
- Department of Immunology and Research Center of Basic Medical Science; Tianjin Key Laboratory of Cellular and Molecular Immunology; Key Laboratory of Immune Microenvironment and Diseases, Ministry of Education of China, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Qi Xiang Tai Road No.22, Tianjin, 300070, China.
| | - Huan Zhang
- Department of Immunology and Research Center of Basic Medical Science; Tianjin Key Laboratory of Cellular and Molecular Immunology; Key Laboratory of Immune Microenvironment and Diseases, Ministry of Education of China, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Qi Xiang Tai Road No.22, Tianjin, 300070, China.
| | - Zhenyi Xue
- Department of Immunology and Research Center of Basic Medical Science; Tianjin Key Laboratory of Cellular and Molecular Immunology; Key Laboratory of Immune Microenvironment and Diseases, Ministry of Education of China, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Qi Xiang Tai Road No.22, Tianjin, 300070, China.
| | - Yurong Da
- Department of Immunology and Research Center of Basic Medical Science; Tianjin Key Laboratory of Cellular and Molecular Immunology; Key Laboratory of Immune Microenvironment and Diseases, Ministry of Education of China, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Qi Xiang Tai Road No.22, Tianjin, 300070, China.
| | - Ning Zhang
- Department of Immunology and Research Center of Basic Medical Science; Tianjin Key Laboratory of Cellular and Molecular Immunology; Key Laboratory of Immune Microenvironment and Diseases, Ministry of Education of China, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Qi Xiang Tai Road No.22, Tianjin, 300070, China.
| | - Yongcheng Hu
- Department of Orthopaedic Oncology, Tianjin Hospital, Tianjin, China.
| | - Zhi Yao
- Department of Immunology and Research Center of Basic Medical Science; Tianjin Key Laboratory of Cellular and Molecular Immunology; Key Laboratory of Immune Microenvironment and Diseases, Ministry of Education of China, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Qi Xiang Tai Road No.22, Tianjin, 300070, China.
| | - Liang Qiao
- Storr Liver Unit, Westmead Millennium Institute, The Western Clinical School of the University of Sydney, Westmead, NSW, Australia.
| | - Fei Gao
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Rongxin Zhang
- Department of Immunology and Research Center of Basic Medical Science; Tianjin Key Laboratory of Cellular and Molecular Immunology; Key Laboratory of Immune Microenvironment and Diseases, Ministry of Education of China, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Qi Xiang Tai Road No.22, Tianjin, 300070, China.
| |
Collapse
|
675
|
Tooi M, Komaki M, Morioka C, Honda I, Iwasaki K, Yokoyama N, Ayame H, Izumi Y, Morita I. Placenta Mesenchymal Stem Cell Derived Exosomes Confer Plasticity on Fibroblasts. J Cell Biochem 2016; 117:1658-70. [PMID: 26640165 DOI: 10.1002/jcb.25459] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 12/04/2015] [Indexed: 12/22/2022]
Abstract
Mesenchymal stem cell (MSC)-conditioned medium (MSC-CM) has been reported to enhance wound healing. Exosomes contain nucleic acids, proteins, and lipids, and function as an intercellular communication vehicle for mediating some paracrine effects. However, the function of MSC-derived exosomes (MSC-exo) remains elusive. In this study, we isolated human placenta MSC (PlaMSC)-derived exosomes (PlaMSC-exo) and examined their function in vitro. PlaMSCs were isolated from human term placenta using enzymatic digestion. PlaMSC-exo were prepared from the conditioned medium of PlaMSC (PlaMSC-CM) by ultracentrifugation. The expression of stemness-related genes, such as OCT4 and NANOG, in normal adult human dermal fibroblasts (NHDF) after incubation with PlaMSC-exo was measured by real-time reverse transcriptase PCR analysis (real-time PCR). The effect of PlaMSC-exo on OCT4 transcription activity was assessed using Oct4-EGFP reporter mice-derived dermal fibroblasts. The stimulating effects of PlaMSC-exo on osteoblastic and adipocyte-differentiation of NHDF were evaluated by alkaline phosphatase (ALP), and Alizarin red S- and oil red O-staining, respectively. The expression of osteoblast- and adipocyte-related genes was also assessed by real-time PCR. The treatment of NHDF with PlaMSC-exo significantly upregulated OCT4 and NANOG mRNA expression. PlaMSC-exo also enhanced OCT4 transcription. The NHDF treated with PlaMSC-exo exhibited osteoblastic and adipocyte-differentiation in osteogenic and adipogenic induction media. PlaMSC-exo increase the expression of OCT4 and NANOG mRNA in fibroblasts. As a result, PlaMSC-exo influence the differentiation competence of fibroblasts to both osteoblastic and adipocyte-differentiation. It shows a new feature of MSCs and the possibility of clinical application of MSC-exo. J. Cell. Biochem. 117: 1658-1670, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Masayuki Tooi
- Department of Periodontology, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Motohiro Komaki
- Department of Nanomedicine (DNP), Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Chikako Morioka
- Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Izumi Honda
- Department of Comprehensive Reproductive Medicine, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Kengo Iwasaki
- Department of Nanomedicine (DNP), Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Naoki Yokoyama
- Life Science Laboratory, Research and Development Center, Dai Nippon Printing Co., Ltd., 1-1-1 Kaga-cho, Shinjuku-ku, Tokyo, 162-8001, Japan
| | - Hirohito Ayame
- Life Science Laboratory, Research and Development Center, Dai Nippon Printing Co., Ltd., 1-1-1 Kaga-cho, Shinjuku-ku, Tokyo, 162-8001, Japan
| | - Yuichi Izumi
- Department of Periodontology, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Ikuo Morita
- Department of Cellular Physiological Chemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan
| |
Collapse
|
676
|
Kamprom W, Kheolamai P, U-Pratya Y, Supokawej A, Wattanapanitch M, Laowtammathron C, Issaragrisil S. Effects of mesenchymal stem cell-derived cytokines on the functional properties of endothelial progenitor cells. Eur J Cell Biol 2016; 95:153-63. [PMID: 26899034 DOI: 10.1016/j.ejcb.2016.02.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 12/28/2015] [Accepted: 02/03/2016] [Indexed: 01/10/2023] Open
Abstract
Human mesenchymal stem cell (hMSC) is a potential source for cell therapy due to its property to promote tissue repair. Although, it has been known that hMSCs promote tissue repair via angiogenic cytokines, the interaction between hMSC-derived cytokines and the endothelial progenitor cells (EPCs), which play an important role in tissue neovascularization, is poorly characterized. We investigate the effect of cytokine released from different sources of hMSCs including bone marrow and gestational tissues on the EPC functions in vitro. The migration, extracellular matrix invasion and vessel formation of EPCs were studied in the presence or absence of cytokines released from various sources of hMSCs using transwell culture system. The migration of EPCs was highest when co-culture with secretory factors from placenta-derived hMSCs (PL-hMSCs) compared to those co-culture with other sources of hMSCs. For invasion and vessel formation, secretory factors from bone marrow-derived hMSCs (BM-hMSCs) could produce the maximal enhancement compared to other sources. We further identified the secreted cytokines and found that the migratory-enhancing cytokine from PL-hMSCs was PDGF-BB while the enhancing cytokine from BM-hMSCs on invasion was IGF-1. For vessel formation, the cytokines released from BM-hMSCs were IGF1 and SDF-1. In conclusion, hMSCs can release angiogenic cytokines which increase the migration, invasion and vessel forming capacity of EPCs. We can then use hMSCs as a source of angiogenic cytokines to induce neovascularization in injured/ischemic tissues.
Collapse
Affiliation(s)
- Witchayaporn Kamprom
- Department of Immunology, Faculty of Medicine Siriraj hospital, Mahidol University, Bangkok, Thailand; Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Pakpoom Kheolamai
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Division of Cell Biology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand; Center of Excellence in Stem Cell Research, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Yaowalak U-Pratya
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | | | - Methichit Wattanapanitch
- Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Chuti Laowtammathron
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Surapol Issaragrisil
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
677
|
Kopaczka K, Skowron K, Kolanko E, Czekaj P. The relationship between amniotic epithelial cells and their microenvironment. J Appl Biomed 2016. [DOI: 10.1016/j.jab.2015.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
678
|
Buitinga M, Janeczek Portalska K, Cornelissen DJ, Plass J, Hanegraaf M, Carlotti F, de Koning E, Engelse M, van Blitterswijk C, Karperien M, van Apeldoorn A, de Boer J. Coculturing Human Islets with Proangiogenic Support Cells to Improve Islet Revascularization at the Subcutaneous Transplantation Site. Tissue Eng Part A 2016; 22:375-85. [PMID: 26871862 DOI: 10.1089/ten.tea.2015.0317] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
While subcutaneous tissue has been proposed as a clinically relevant site for pancreatic islet transplantation, a major issue of concern remains, which is its poor vascular state. In an effort to overcome this limitation, we present an efficient and reproducible method to form human composite islets (CIs) with proangiogenic cell types in a controlled manner using nonadherent agarose microwell templates. In this study, we assessed the three-dimensional structure, function, and angiogenic potential of human CIs with human mesenchymal stromal cells (hMSCs), with or without human umbilical vein endothelial cells (HUVECs), and preconditioned hMSCs (PC-hMSCs) in EGM-2 under shear stress. Distinct cellular rearrangements could be observed in CIs, but islet functionality was maintained. In vitro angiogenesis assays found significantly enhanced sprout formation in case of CIs. In particular, the number of sprouts emanating from CIs with PC-hMSCs was significantly increased compared to other conditions. Subsequent in vivo assessment confirmed the proangiogenic potential of CIs. However, in contrast to our in vitro angiogenesis assays, CIs with hMSCs and HUVECs exhibited a higher in vivo angiogenic potential compared to control islets or islets combined with hMSCs or PC-hMSCs. These findings highlight the importance and necessity of verifying in vitro studies with in vivo models to reliably predict, in this case, revascularization outcomes. Regardless, we demonstrate here the therapeutic potential of CIs with proangiogenic support cells to enhance islet revascularization at a clinically relevant, although poorly vascularized, transplantation site.
Collapse
Affiliation(s)
- Mijke Buitinga
- 1 Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, The Netherlands
| | - Karolina Janeczek Portalska
- 2 Department of Tissue Regeneration, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, The Netherlands
| | - Dirk-Jan Cornelissen
- 1 Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, The Netherlands
| | - Jacqueline Plass
- 1 Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, The Netherlands
| | - Maaike Hanegraaf
- 3 Department of Nephrology, Leiden University Medical Center , Leiden, The Netherlands
| | - Françoise Carlotti
- 3 Department of Nephrology, Leiden University Medical Center , Leiden, The Netherlands
| | - Eelco de Koning
- 3 Department of Nephrology, Leiden University Medical Center , Leiden, The Netherlands .,4 Department of Endocrinology, Leiden University Medical Center , Leiden, The Netherlands .,5 Hubrecht Institute-Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht , Utrecht, The Netherlands
| | - Marten Engelse
- 3 Department of Nephrology, Leiden University Medical Center , Leiden, The Netherlands
| | - Clemens van Blitterswijk
- 6 Department of Complex Tissue Regeneration, Institute for Technology Inspired Regenerative Medicine (MERLN), Maastricht University , Maastricht, The Netherlands
| | - Marcel Karperien
- 1 Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, The Netherlands
| | - Aart van Apeldoorn
- 1 Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente , Enschede, The Netherlands
| | - Jan de Boer
- 7 Laboratory for Cell Biology-Inspired Tissue Engineering, Institute for Technology Inspired Regenerative Medicine (MERLN) , Maastricht, The Netherlands
| |
Collapse
|
679
|
Miceli M, Dell'Aversana C, Russo R, Rega C, Cupelli L, Ruvo M, Altucci L, Chambery A. Secretome profiling of cytokines and growth factors reveals that neuro-glial differentiation is associated with the down-regulation of Chemokine Ligand 2 (MCP-1/CCL2) in amniotic fluid derived-mesenchymal progenitor cells. Proteomics 2016; 16:674-88. [PMID: 26604074 DOI: 10.1002/pmic.201500223] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 10/09/2015] [Accepted: 11/20/2015] [Indexed: 01/09/2023]
Abstract
Secreted cytokines and growth factors play a key role in the modulation of stem cell proliferation, differentiation and survival. To investigate the interplay between the changes in their expression levels, we used the newly characterized human amniotic fluid derived-mesenchymal progenitor MePR-2B cell line differentiated to a neuro-glial phenotype and exploited the very high sensitivity and versatility of magnetic beads-based immunoassays. We found that a sub-set of proteins, including the cytokines IL-6, TNFα, IL-15, IFNγ, IL-8, IL-1ra, MCP-1/CCL2, RANTES and the growth factor PDGFbb, underwent a significant down-regulation following neuro-glial differentiation, whereas the expression levels of IL-12 p70, IL-5, IL-7, bFGF, VEGF and G-CSF were increased. The role of MCP-1/CCL2, previously identified as a regulator of neural progenitor stem cell differentiation, has been further investigated at transcriptional level, revealing that both the chemokine and its receptor are co-expressed in MePR-2B cells and that are regulated upon differentiation, suggesting the presence of an autocrine and paracrine loop in differentiating cells. Moreover, we demonstrated that exogenous CCL2 is capable to affect neuro-glial differentiation in MePR-2B cells, thus providing novel evidences for the potential involvement of chemokine-mediated signaling in progenitor/stem cells differentiation processes and fate specification.
Collapse
Affiliation(s)
- Marco Miceli
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Seconda Università di Napoli, Napoli, Italy.,Istituto di Genetica e Biofisica 'Adriano Buzzati Traverso' IGB, CNR, Napoli, Italy
| | - Carmela Dell'Aversana
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Seconda Università di Napoli, Napoli, Italy.,Istituto di Genetica e Biofisica 'Adriano Buzzati Traverso' IGB, CNR, Napoli, Italy
| | - Rosita Russo
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Seconda Università degli Studi di Napoli, Caserta, Italy
| | - Camilla Rega
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Seconda Università degli Studi di Napoli, Caserta, Italy
| | - Lorenzo Cupelli
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Seconda Università di Napoli, Napoli, Italy.,Istituto di Genetica e Biofisica 'Adriano Buzzati Traverso' IGB, CNR, Napoli, Italy
| | - Menotti Ruvo
- Istituto di Biostrutture e Bioimmagini, IBB, CNR, Napoli, Italy
| | - Lucia Altucci
- Dipartimento di Biochimica, Biofisica e Patologia Generale, Seconda Università di Napoli, Napoli, Italy.,Istituto di Genetica e Biofisica 'Adriano Buzzati Traverso' IGB, CNR, Napoli, Italy
| | - Angela Chambery
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Seconda Università degli Studi di Napoli, Caserta, Italy.,IRCCS, Multimedica, Milano, Italy
| |
Collapse
|
680
|
Yaochite JNU, de Lima KWA, Caliari-Oliveira C, Palma PVB, Couri CEB, Simões BP, Covas DT, Voltarelli JC, Oliveira MC, Donadi EA, Malmegrim KCR. Multipotent mesenchymal stromal cells from patients with newly diagnosed type 1 diabetes mellitus exhibit preserved in vitro and in vivo immunomodulatory properties. Stem Cell Res Ther 2016; 7:14. [PMID: 26781648 PMCID: PMC4861132 DOI: 10.1186/s13287-015-0261-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 11/16/2015] [Accepted: 12/07/2015] [Indexed: 12/25/2022] Open
Abstract
Background Type 1 diabetes mellitus (T1D) is characterized by autoimmune responses resulting in destruction of insulin-producing pancreatic beta cells. Multipotent mesenchymal stromal cells (MSCs) exhibit immunomodulatory potential, migratory capacity to injured areas and may contribute to tissue regeneration by the secretion of bioactive factors. Therefore, MSCs are considered as a promising approach to treat patients with different autoimmune diseases (AID), including T1D patients. Phenotypical and functional alterations have been reported in MSCs derived from patients with different AID. However, little is known about the properties of MSCs derived from patients with T1D. Since autoimmunity and the diabetic microenvironment may affect the biology of MSCs, it becomes important to investigate whether these cells are suitable for autologous transplantation. Thus, the aim of the present study was to evaluate the in vitro properties and the in vivo therapeutic efficacy of MSCs isolated from bone marrow of newly diagnosed T1D patients (T1D-MSCs) and to compare them with MSCs from healthy individuals (C-MSCs). Methods T1D-MSCs and C-MSCs were isolated and cultured until third passage. Then, morphology, cell diameter, expression of surface markers, differentiation potential, global microarray analyses and immunosuppressive capacity were in vitro analyzed. T1D-MSCs and C-MSCs therapeutic potential were evaluated using a murine experimental model of streptozotocin (STZ)-induced diabetes. Results T1D-MSCs and C-MSCs presented similar morphology, immunophenotype, differentiation potential, gene expression of immunomodulatory molecules and in vitro immunosuppressive capacity. When administered into diabetic mice, both T1D-MSCs and C-MSCs were able to reverse hyperglycemia, improve beta cell function and modulate pancreatic cytokine levels. Conclusions Thus, bone marrow MSCs isolated from T1D patients recently after diagnosis are not phenotypically or functionally impaired by harmful inflammatory and metabolic diabetic conditions. Our results provide support for the use of autologous MSCs for treatment of newly diagnosed T1D patients. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0261-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Juliana Navarro Ueda Yaochite
- Department of Biochemistry and Immunology, Basic and Applied Immunology Program, School of Medicine of Ribeirão Preto, University of São Paulo, Av. Bandeirantes, 3900, Monte Alegre, 14049-900, Ribeirão Preto, São Paulo, Brazil. .,Department of Clinical and Toxicological Analysis, Federal University of Ceará, Alexandre Baraúna 949, Rodolfo Teófilo, 60430-160, Fortaleza, Ceará, Brazil. .,Regional Blood Center of Ribeirão Preto, University of São Paulo, Tenente Catão Roxo, 2501, Monte Alegre, 14051-140, Ribeirão Preto, São Paulo, Brazil.
| | - Kalil Willian Alves de Lima
- Department of Biochemistry and Immunology, Basic and Applied Immunology Program, School of Medicine of Ribeirão Preto, University of São Paulo, Av. Bandeirantes, 3900, Monte Alegre, 14049-900, Ribeirão Preto, São Paulo, Brazil.
| | - Carolina Caliari-Oliveira
- Department of Biochemistry and Immunology, Basic and Applied Immunology Program, School of Medicine of Ribeirão Preto, University of São Paulo, Av. Bandeirantes, 3900, Monte Alegre, 14049-900, Ribeirão Preto, São Paulo, Brazil. .,Regional Blood Center of Ribeirão Preto, University of São Paulo, Tenente Catão Roxo, 2501, Monte Alegre, 14051-140, Ribeirão Preto, São Paulo, Brazil.
| | - Patricia Vianna Bonini Palma
- Regional Blood Center of Ribeirão Preto, University of São Paulo, Tenente Catão Roxo, 2501, Monte Alegre, 14051-140, Ribeirão Preto, São Paulo, Brazil.
| | - Carlos Eduardo Barra Couri
- Department of Clinical Medicine, School of Medicine of Ribeirão Preto, University of São Paulo, Tenente Catão Roxo, 2501, Monte Alegre, 14051-140, Ribeirão Preto, São Paulo, Brazil.
| | - Belinda Pinto Simões
- Department of Clinical Medicine, School of Medicine of Ribeirão Preto, University of São Paulo, Tenente Catão Roxo, 2501, Monte Alegre, 14051-140, Ribeirão Preto, São Paulo, Brazil.
| | - Dimas Tadeu Covas
- Regional Blood Center of Ribeirão Preto, University of São Paulo, Tenente Catão Roxo, 2501, Monte Alegre, 14051-140, Ribeirão Preto, São Paulo, Brazil. .,Department of Clinical Medicine, School of Medicine of Ribeirão Preto, University of São Paulo, Tenente Catão Roxo, 2501, Monte Alegre, 14051-140, Ribeirão Preto, São Paulo, Brazil.
| | | | - Maria Carolina Oliveira
- Department of Clinical Medicine, School of Medicine of Ribeirão Preto, University of São Paulo, Tenente Catão Roxo, 2501, Monte Alegre, 14051-140, Ribeirão Preto, São Paulo, Brazil.
| | - Eduardo Antônio Donadi
- Department of Biochemistry and Immunology, Basic and Applied Immunology Program, School of Medicine of Ribeirão Preto, University of São Paulo, Av. Bandeirantes, 3900, Monte Alegre, 14049-900, Ribeirão Preto, São Paulo, Brazil. .,Department of Clinical Medicine, School of Medicine of Ribeirão Preto, University of São Paulo, Tenente Catão Roxo, 2501, Monte Alegre, 14051-140, Ribeirão Preto, São Paulo, Brazil.
| | - Kelen Cristina Ribeiro Malmegrim
- Regional Blood Center of Ribeirão Preto, University of São Paulo, Tenente Catão Roxo, 2501, Monte Alegre, 14051-140, Ribeirão Preto, São Paulo, Brazil. .,Department of Clinical, Toxicological and Bromatological Analysis, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café, Monte Alegre, 14040-903, Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
681
|
Gohil SV, Kuo C, Adams DJ, Maye P, Rowe DW, Nair LS. Evaluation of the donor cell contribution in rh
BMP
‐2 mediated bone formation with chitosan thermogels using fluorescent protein reporter mice. J Biomed Mater Res A 2016; 104:928-41. [DOI: 10.1002/jbm.a.35634] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 11/18/2015] [Accepted: 12/18/2015] [Indexed: 12/13/2022]
Affiliation(s)
- Shalini V. Gohil
- Department of Orthopaedic SurgeryUConn HealthFarmington Connecticut06030
- Institute for Regenerative Engineering, The Raymond Beverly Sackler Center for Biomedical, Biological, Physical and Engineering SciencesUConn HealthFarmington Connecticut06030
| | - Chia‐Ling Kuo
- Connecticut Institute for Clinical and Translational Science, Institute for Systems Genomics, University of ConnecticutFarmington Connecticut06030
| | - Douglas J. Adams
- Department of Orthopaedic SurgeryUConn HealthFarmington Connecticut06030
| | - Peter Maye
- Center for Regenerative Medicine and Skeletal Development, Department of Reconstructive Sciences, School of Dental MedicineUConn HealthFarmington Connecticut06030
| | - David W. Rowe
- Center for Regenerative Medicine and Skeletal Development, Department of Reconstructive Sciences, School of Dental MedicineUConn HealthFarmington Connecticut06030
| | - Lakshmi S. Nair
- Department of Orthopaedic SurgeryUConn HealthFarmington Connecticut06030
- Institute for Regenerative Engineering, The Raymond Beverly Sackler Center for Biomedical, Biological, Physical and Engineering SciencesUConn HealthFarmington Connecticut06030
- Departments of Material Science and Engineering, Biomedical Engineering and Institute of Material ScienceUniversity of ConnecticutStorrs Connecticut06269
| |
Collapse
|
682
|
L Ramos T, Sánchez-Abarca LI, Muntión S, Preciado S, Puig N, López-Ruano G, Hernández-Hernández Á, Redondo A, Ortega R, Rodríguez C, Sánchez-Guijo F, del Cañizo C. MSC surface markers (CD44, CD73, and CD90) can identify human MSC-derived extracellular vesicles by conventional flow cytometry. Cell Commun Signal 2016; 14:2. [PMID: 26754424 PMCID: PMC4709865 DOI: 10.1186/s12964-015-0124-8] [Citation(s) in RCA: 224] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 12/21/2015] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Human mesenchymal stromal cells (hMSC) are multipotent cells with both regenerative and immunomodulatory activities making them an attractive tool for cellular therapy. In the last few years it has been shown that the beneficial effects of hMSC may be due to paracrine effects and, at least in part, mediated by extracellular vesicles (EV). EV have emerged as important mediators of cell-to-cell communication. Flow cytometry (FCM) is a routine technology used in most clinical laboratories and could be used as a methodology for hMSC-EV characterization. Although several reports have characterized EV by FCM, a specific panel and protocol for hMSC-derived EV is lacking. The main objective of our study was the characterization of hMSC-EV using a standard flow cytometer. METHODS Human MSC from bone marrow of healthy donors, mesenchymal cell lines (HS-5 and hTERT) and a leukemic cell line (K562 cells) were used to obtain EV for FCM characterization. EV released from the different cell lines were isolated by ultracentrifugation and were characterized, using a multi-parametric analysis, in a conventional flow cytometer. EV characterization by transmission electron microscopy (TEM), western blot (WB) and Nano-particle tracking analysis (NTA) was also performed. RESULTS EV membranes are constituted by the combination of specific cell surface molecules depending on their cell of origin, together with specific proteins like tetraspanins (e.g. CD63). We have characterized by FCM the EV released from BM-hMSC, that were defined as particles less than 0.9 μm, positive for the hMSC markers (CD90, CD44 and CD73) and negative for CD34 and CD45 (hematopoietic markers). In addition, hMSC-derived EV were also positive for CD63 and CD81, the two characteristic markers of EV. To validate our characterization strategy, EV from mesenchymal cell lines (hTERT/HS-5) were also studied, using the leukemia cell line (K562) as a negative control. EV released from mesenchymal cell lines displayed the same immunophenotypic profile as the EV from primary BM-hMSC, while the EV derived from K562 cells did not show hMSC markers. We further validated the panel using EV from hMSC transduced with GFP. Finally, EV derived from the different sources (hMSC, hTERT/HS-5 and K562) were also characterized by WB, TEM and NTA, demonstrating the expression by WB of the exosomal markers CD63 and CD81, as well as CD73 in those from MSC origin. EV morphology and size/concentration was confirmed by TEM and NTA, respectively. CONCLUSION We described a strategy that allows the identification and characterization by flow cytometry of hMSC-derived EV that can be routinely used in most laboratories with a standard flow cytometry facility.
Collapse
Affiliation(s)
- Teresa L Ramos
- Servicio de Hematología, IBSAL-Hospital Universitario de Salamanca, Paseo de San Vicente 58-182, 37007, Salamanca, Spain. .,Centro de Investigación del Cáncer, Universidad de Salamanca, Salamanca, Spain. .,Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, León, Spain. .,Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain.
| | - Luis Ignacio Sánchez-Abarca
- Servicio de Hematología, IBSAL-Hospital Universitario de Salamanca, Paseo de San Vicente 58-182, 37007, Salamanca, Spain. .,Centro de Investigación del Cáncer, Universidad de Salamanca, Salamanca, Spain. .,Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, León, Spain. .,Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain.
| | - Sandra Muntión
- Servicio de Hematología, IBSAL-Hospital Universitario de Salamanca, Paseo de San Vicente 58-182, 37007, Salamanca, Spain. .,Centro de Investigación del Cáncer, Universidad de Salamanca, Salamanca, Spain. .,Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, León, Spain. .,Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain.
| | - Silvia Preciado
- Servicio de Hematología, IBSAL-Hospital Universitario de Salamanca, Paseo de San Vicente 58-182, 37007, Salamanca, Spain. .,Centro de Investigación del Cáncer, Universidad de Salamanca, Salamanca, Spain. .,Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, León, Spain. .,Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain.
| | - Noemí Puig
- Servicio de Hematología, IBSAL-Hospital Universitario de Salamanca, Paseo de San Vicente 58-182, 37007, Salamanca, Spain. .,Centro de Investigación del Cáncer, Universidad de Salamanca, Salamanca, Spain. .,Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, León, Spain. .,Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain.
| | - Guillermo López-Ruano
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain. .,Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Salamanca, Spain.
| | - Ángel Hernández-Hernández
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain. .,Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Salamanca, Spain.
| | - Alba Redondo
- Servicio de Hematología, IBSAL-Hospital Universitario de Salamanca, Paseo de San Vicente 58-182, 37007, Salamanca, Spain. .,Centro de Investigación del Cáncer, Universidad de Salamanca, Salamanca, Spain. .,Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, León, Spain. .,Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain.
| | - Rebeca Ortega
- Servicio de Hematología, IBSAL-Hospital Universitario de Salamanca, Paseo de San Vicente 58-182, 37007, Salamanca, Spain.
| | - Concepción Rodríguez
- Servicio de Hematología, IBSAL-Hospital Universitario de Salamanca, Paseo de San Vicente 58-182, 37007, Salamanca, Spain. .,Centro de Investigación del Cáncer, Universidad de Salamanca, Salamanca, Spain. .,Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, León, Spain. .,Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain.
| | - Fermín Sánchez-Guijo
- Servicio de Hematología, IBSAL-Hospital Universitario de Salamanca, Paseo de San Vicente 58-182, 37007, Salamanca, Spain. .,Centro de Investigación del Cáncer, Universidad de Salamanca, Salamanca, Spain. .,Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, León, Spain. .,Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain.
| | - Consuelo del Cañizo
- Servicio de Hematología, IBSAL-Hospital Universitario de Salamanca, Paseo de San Vicente 58-182, 37007, Salamanca, Spain. .,Centro de Investigación del Cáncer, Universidad de Salamanca, Salamanca, Spain. .,Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, León, Spain. .,Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain.
| |
Collapse
|
683
|
Murrell WD, Anz AW, Badsha H, Bennett WF, Boykin RE, Caplan AI. Regenerative treatments to enhance orthopedic surgical outcome. PM R 2016; 7:S41-S52. [PMID: 25864660 DOI: 10.1016/j.pmrj.2015.01.015] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 01/09/2015] [Accepted: 01/14/2015] [Indexed: 12/20/2022]
Abstract
In orthopedic surgery there has been a never-ending quest to improve surgical outcome and the patient's experience. Progression has been marked by the refinement of surgical techniques and instruments and later by enhanced diagnostic imaging capability, specifically magnetic resonance. Over time implant optimization was achieved, along with the development of innovative minimally invasive arthroscopic technical skills to leverage new versions of classic procedures and implants to improve short-term patient morbidity and initial, mid-term, and long-term patient outcomes. The use of regenerative and/or biological adjuncts to aid the healing process has followed in the drive for continual improvement, and major breakthroughs in basic science have significantly unraveled the mechanisms of key healing and regenerative pathways. A wide spectrum of primary and complementary regenerative treatments is becoming increasingly available, including blood-derived preparations, growth factors, bone marrow preparations, and stem cells. This is a new era in the application of biologically active material, and it is transforming clinical practice by providing effective supportive treatments either at the time of the index procedure or during the postoperative period. Regenerative treatments are currently in active use to enhance many areas of orthopedic surgery in an attempt to improve success and outcome. In this review we provide a comprehensive overview of the peer-reviewed evidence-based literature, highlighting the clinical outcomes in humans both with preclinical data and human clinical trials involving regenerative preparations within the areas of rotator cuff, meniscus, ligament, and articular cartilage surgical repair.
Collapse
Affiliation(s)
- William D Murrell
- Dr Humeira Badsha Medical Center, Dubai, United Arab Emirates 391203; and Fort Belvoir Community Hospital, Department of Orthopaedics, Podiatry, Physical Therapy, and Rehabilitation, Ft. Belvoir, VVA 22060
| | - Adam W Anz
- Andrews Reseach & Education Institute, Gulf Breeze, FL
| | - Humeira Badsha
- Dr Humeira Badsha Medical Center, Dubai, United Arab Emirates 391203
| | | | | | - Arnold I Caplan
- Department of Biology, Skeletal Research Center, Case Western Reserve University, Cleveland, OH
| |
Collapse
|
684
|
Broekman W, Amatngalim GD, de Mooij-Eijk Y, Oostendorp J, Roelofs H, Taube C, Stolk J, Hiemstra PS. TNF-α and IL-1β-activated human mesenchymal stromal cells increase airway epithelial wound healing in vitro via activation of the epidermal growth factor receptor. Respir Res 2016; 17:3. [PMID: 26753875 PMCID: PMC4710048 DOI: 10.1186/s12931-015-0316-1] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 12/15/2015] [Indexed: 12/13/2022] Open
Abstract
Background Mesenchymal stromal cells (MSCs) are investigated for their potential to reduce inflammation and to repair damaged tissue. Inflammation and tissue damage are hallmarks of chronic obstructive pulmonary disease (COPD) and MSC infusion is a promising new treatment for COPD. Inflammatory mediators attract MSCs to sites of inflammation and affect their immune-modulatory properties, but little is known about their effect on regenerative properties of MSCs. This study investigates the effect of the pro-inflammatory cytokines TNF-α and IL-1β on the regenerative potential of MSCs, using an in vitro wound healing model of airway epithelial cells. Methods Standardized circular wounds were created by scraping cultures of the airway epithelial cell line NCI-H292 and primary bronchial epithelial cells cultured at the air-liquid interface (ALI-PBEC), and subsequently incubated with MSC conditioned medium (MSC-CM) that was generated in presence or absence of TNF-α/IL-1β. Remaining wound size was measured up to 72 h. Phosphorylation of ERK1/2 by MSC-CM was assessed using Western blot. Inhibitors for EGFR and c-Met signaling were used to investigate the contribution of these receptors to wound closure and to ERK1/2 phosphorylation. Transactivation of EGFR by MSC-CM was investigated using a TACE inhibitor, and RT-PCR was used to quantify mRNA expression of several growth factors in MSCs and NCI-H292. Results Stimulation of MSCs with the pro-inflammatory cytokines TNF-α and IL-1β increased the mRNA expression of various growth factors by MCSs and enhanced the regenerative potential of MSCs in an in vitro model of airway epithelial injury using NCI-H292 airway epithelial cells. Conditioned medium from cytokine stimulated MSCs induced ERK1/2 phosphorylation in NCI-H292, predominantly via EGFR; it induced ADAM-mediated transactivation of EGFR, and it induced airway epithelial expression of several EGFR ligands. The contribution of activation of c-Met via HGF to increased repair could not be confirmed by inhibitor experiments. Conclusion Our data imply that at sites of tissue damage, when inflammatory mediators are present, for example in lungs of COPD patients, MSCs become more potent inducers of repair, in addition to their well-known immune-modulatory properties.
Collapse
Affiliation(s)
- Winifred Broekman
- Department of Pulmonology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| | - Gimano D Amatngalim
- Department of Pulmonology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| | - Yvonne de Mooij-Eijk
- Department of Pulmonology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| | - Jaap Oostendorp
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands.
| | - Helene Roelofs
- Department of Immunohaematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands.
| | - Christian Taube
- Department of Pulmonology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| | - Jan Stolk
- Department of Pulmonology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| |
Collapse
|
685
|
Endothelial Progenitor Cell Migration-Enhancing Factors in the Secretome of Placental-Derived Mesenchymal Stem Cells. Stem Cells Int 2016; 2016:2514326. [PMID: 26880942 PMCID: PMC4736766 DOI: 10.1155/2016/2514326] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 10/13/2015] [Accepted: 10/25/2015] [Indexed: 01/12/2023] Open
Abstract
Therapeutic potentials of mesenchymal stem cells (MSCs) depend largely on their ability to secrete cytokines or factors that modulate immune response, enhance cell survival, and induce neovascularization in the target tissues. We studied the secretome profile of gestational tissue-derived MSCs and their effects on functions of endothelial progenitor cells (EPCs), another angiogenic cell type that plays an important role during the neovascularization. MSCs derived from placental tissues (PL-MSCs) significantly enhanced EPC migration while BM-MSCs, which are the standard source of MSCs for various clinical applications, did not. By using protein fractionation and mass spectrometry analysis, we identified several novel candidates for EPC migration enhancing factor in PL-MSCs secretome that could be used to enhance neovascularization in the injured/ischemic tissues. We recommend that the strategy developed in our study could be used to systematically identify therapeutically useful molecules in the secretomes of other MSC sources for the clinical applications.
Collapse
|
686
|
Neuromuscular Regeneration: Perspective on the Application of Mesenchymal Stem Cells and Their Secretion Products. Stem Cells Int 2016; 2016:9756973. [PMID: 26880998 PMCID: PMC4736584 DOI: 10.1155/2016/9756973] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 10/12/2015] [Accepted: 11/16/2015] [Indexed: 02/08/2023] Open
Abstract
Mesenchymal stem cells are posing as a promising character in the most recent therapeutic strategies and, since their discovery, extensive knowledge on their features and functions has been gained. In recent years, innovative sources have been disclosed in alternative to the bone marrow, conveying their associated ethical concerns and ease of harvest, such as the umbilical cord tissue and the dental pulp. These are also amenable of cryopreservation and thawing for desired purposes, in benefit of the donor itself or other patients in pressing need. These sources present promising possibilities in becoming useful cell sources for therapeutic applications in the forthcoming years. Effective and potential applications of these cellular-based strategies for the regeneration of peripheral nerve are overviewed, documenting recent advances and identified issues for this research area in the near future. Finally, besides the differentiation capacities attributed to mesenchymal stem cells, advances in the recognition of their effective mode of action in the regenerative theatre have led to a new area of interest: the mesenchymal stem cells' secretome. The paracrine modulatory pathway appears to be a major mechanism by which these are beneficial to nerve regeneration and comprehension on the specific growth factors, cytokine, and extracellular molecules secretion profiles is therefore of great interest.
Collapse
|
687
|
AHN MJ, JEONG SG, CHO GW. Antisenescence activity of G9a inhibitor BIX01294 on human bone marrow mesenchymal stromal cells. Turk J Biol 2016. [DOI: 10.3906/biy-1507-11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
688
|
Abstract
Acute radiation syndrome affects military personnel and civilians following the uncontrolled dispersal of radiation, such as that caused by detonation of nuclear devices and inappropriate medical treatments. Therefore, there is a growing need for medical interventions that facilitate the improved recovery of victims and patients. One promising approach may be cell therapy, which, when appropriately implemented, may facilitate recovery from whole body injuries. This editorial highlights the current knowledge regarding the use of mesenchymal stem cells for the treatment of acute radiation syndrome, the benefits and limitations of which are under investigation. Establishing successful therapies for acute radiation syndrome may require using such a therapeutic approach in addition to conventional approaches.
Collapse
Affiliation(s)
- Risaku Fukumoto
- Educational-Scientific Center, Faculty of Health Sciences, Medical University of Białystok, ul. Szpitalna 37, 15-295 Białystok, Poland
| |
Collapse
|
689
|
Song K, Yang Y, Wu S, Zhang Y, Feng S, Wang H, Wang Y, Wang L, Liu T. In vitro culture and harvest of BMMSCs on the surface of a novel thermosensitive glass microcarrier. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 58:324-30. [DOI: 10.1016/j.msec.2015.08.033] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 07/18/2015] [Accepted: 08/22/2015] [Indexed: 12/28/2022]
|
690
|
Stavely R, Robinson AM, Miller S, Boyd R, Sakkal S, Nurgali K. Allogeneic guinea pig mesenchymal stem cells ameliorate neurological changes in experimental colitis. Stem Cell Res Ther 2015; 6:263. [PMID: 26718461 PMCID: PMC4697327 DOI: 10.1186/s13287-015-0254-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 11/12/2015] [Accepted: 12/02/2015] [Indexed: 12/13/2022] Open
Abstract
Background The use of mesenchymal stem cells (MSCs) to treat inflammatory bowel disease (IBD) is of great interest because of their immunomodulatory properties. Damage to the enteric nervous system (ENS) is implicated in IBD pathophysiology and disease progression. The most commonly used model to study inflammation-induced changes to the ENS is 2,4,6-trinitrobenzene-sulfonate acid (TNBS)-induced colitis in guinea pigs; however, no studies using guinea pig MSCs in colitis have been performed. This study aims to isolate and characterise guinea pig MSCs and then test their therapeutic potential for the treatment of enteric neuropathy associated with intestinal inflammation. Methods MSCs from guinea pig bone marrow and adipose tissue were isolated and characterised in vitro. In in vivo experiments, guinea pigs received either TNBS for the induction of colitis or sham treatment by enema. MSCs were administered at a dose of 1 × 106 cells via enema 3 h after the induction of colitis. Colon tissues were collected 24 and 72 h after TNBS administration to assess the level of inflammation and damage to the ENS. The secretion of transforming growth factor-β1 (TGF-β1) was analysed in MSC conditioned medium by flow cytometry. Results Cells isolated from both sources were adherent to plastic, multipotent and expressed some human MSC surface markers. In vitro characterisation revealed distinct differences in growth kinetics, clonogenicity and cell morphology between MSC types. In an in vivo model of TNBS-induced colitis, guinea pig bone marrow MSCs were comparatively more efficacious than adipose tissue MSCs in attenuating weight loss, colonic tissue damage and leukocyte infiltration into the mucosa and myenteric plexus. MSCs from both sources were equally neuroprotective in the amelioration of enteric neuronal loss and changes to the neurochemical coding of neuronal subpopulations. MSCs from both sources secreted TGF-β1 which exerted neuroprotective effects in vitro. Conclusions This study is the first evaluating the functional capacity of guinea pig bone marrow and adipose tissue-derived MSCs and providing evidence of their neuroprotective value in an animal model of colitis. In vitro characteristics of MSCs cannot be extrapolated to their therapeutic efficacy. TGF-β1 released by both types of MSCs might have contributed to the attenuation of enteric neuropathy associated with colitis.
Collapse
Affiliation(s)
- Rhian Stavely
- Centre for Chronic Disease, College of Health and Biomedicine, Western Centre for Health, Research and Education, Sunshine Hospital, 176 Furlong road, Melbourne, 3021, Victoria, Australia.
| | - Ainsley M Robinson
- Centre for Chronic Disease, College of Health and Biomedicine, Western Centre for Health, Research and Education, Sunshine Hospital, 176 Furlong road, Melbourne, 3021, Victoria, Australia.
| | - Sarah Miller
- Centre for Chronic Disease, College of Health and Biomedicine, Western Centre for Health, Research and Education, Sunshine Hospital, 176 Furlong road, Melbourne, 3021, Victoria, Australia.
| | - Richard Boyd
- Department of Anatomy and Developmental Biology, Monash University, 19 Innovation Walk, Clayton, 3800, Victoria, Australia.
| | - Samy Sakkal
- Centre for Chronic Disease, College of Health and Biomedicine, Western Centre for Health, Research and Education, Sunshine Hospital, 176 Furlong road, Melbourne, 3021, Victoria, Australia.
| | - Kulmira Nurgali
- Centre for Chronic Disease, College of Health and Biomedicine, Western Centre for Health, Research and Education, Sunshine Hospital, 176 Furlong road, Melbourne, 3021, Victoria, Australia.
| |
Collapse
|
691
|
The Modulatory Effects of Mesenchymal Stem Cells on Osteoclastogenesis. Stem Cells Int 2015; 2016:1908365. [PMID: 26823668 PMCID: PMC4707367 DOI: 10.1155/2016/1908365] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 09/21/2015] [Indexed: 12/14/2022] Open
Abstract
The effect of mesenchymal stem cells (MSCs) on bone formation has been extensively demonstrated through several in vitro and in vivo studies. However, few studies addressed the effect of MSCs on osteoclastogenesis and bone resorption. Under physiological conditions, MSCs support osteoclastogenesis through producing the main osteoclastogenic cytokines, RANKL and M-CSF. However, during inflammation, MSCs suppress osteoclast formation and activity, partly via secretion of the key anti-osteoclastogenic factor, osteoprotegerin (OPG). In vitro, co-culture of MSCs with osteoclasts in the presence of high concentrations of osteoclast-inducing factors might reflect the in vivo inflammatory pathology and prompt MSCs to exert an osteoclastogenic suppressive effect. MSCs thus seem to have a dual effect, by stimulating or inhibiting osteoclastogenesis, depending on the inflammatory milieu. This effect of MSCs on osteoclast formation seems to mirror the effect of MSCs on other immune cells, and may be exploited for the therapeutic potential of MSCs in bone loss associated inflammatory diseases.
Collapse
|
692
|
Shimomura A, Iizuka-Kogo A, Yamamoto N, Nomura R. A lower volume culture method for obtaining a larger yield of neuron-like cells from mesenchymal stem cells. Med Mol Morphol 2015; 49:119-26. [PMID: 26700227 DOI: 10.1007/s00795-015-0131-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 12/08/2015] [Indexed: 01/21/2023]
Abstract
Mesenchymal stem cells (MSCs) represent a promising cell source for stem cell therapy to replace neurons damaged by neurodegenerative diseases. A system designed for in vitro neuronal differentiation of MSCs is an indispensable technique, which provides MSC-derived functional neurons for cell-replacement therapies and valuable information in pre-clinical research. This study investigated the effects of reducing the volume of neural induction medium on cell viability and neural differentiation of MSCs. When MSCs were differentiated in low volumes of neural induction medium, rather than using the conventional method, the cell density on culture dishes significantly increased. The % cell death, including apoptosis and necrosis, was significantly lower in the lower volume method than in the conventional method. There were no significant differences between the lower volume and conventional methods in the expression levels of the neuronal marker genes. In an analysis of immunostaining for a mature neuronal marker, no significant difference was detected between the media volumes. These findings demonstrate that neuronal induction of MSCs in low volumes of differentiation medium promoted survival during differentiation and resulted in larger numbers of MSC-derived neurons, compared to the conventional method. This novel lower volume method offers both financial and cell-yield advantages over the conventional method.
Collapse
Affiliation(s)
- Atsushi Shimomura
- Department of Communication Disorders, Health Sciences University of Hokkaido School of Psychological Science, 2-5 Ainosato, Kita-ku, Sapporo, Hokkaido, 002-8072, Japan. .,Department of Communication Disorders, Health Sciences University of Hokkaido School of Rehabilitation Sciences, 1757 Kanazawa, Tobetsu, Hokkaido, 061-0293, Japan.
| | - Akiko Iizuka-Kogo
- Department of Anatomy and Cell Biology, Gunma University School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma, 371-8511, Japan
| | - Naoki Yamamoto
- Laboratory of Molecular Biology and Histochemistry, Fujita Health University Joint Research Laboratory, 1-98 Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Ryuji Nomura
- Department of Anatomy I, Fujita Health University School of Medicine, 1-98 Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| |
Collapse
|
693
|
Iser IC, Ceschini SM, Onzi GR, Bertoni APS, Lenz G, Wink MR. Conditioned Medium from Adipose-Derived Stem Cells (ADSCs) Promotes Epithelial-to-Mesenchymal-Like Transition (EMT-Like) in Glioma Cells In vitro. Mol Neurobiol 2015; 53:7184-7199. [DOI: 10.1007/s12035-015-9585-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 11/29/2015] [Indexed: 12/21/2022]
|
694
|
Onishi R, Ohnishi S, Higashi R, Watari M, Yamahara K, Okubo N, Nakagawa K, Katsurada T, Suda G, Natsuizaka M, Takeda H, Sakamoto N. Human Amnion-Derived Mesenchymal Stem Cell Transplantation Ameliorates Dextran Sulfate Sodium-Induced Severe Colitis in Rats. Cell Transplant 2015; 24:2601-14. [DOI: 10.3727/096368915x687570] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are a valuable cell source in regenerative medicine. Recently, several studies have shown that MSCs can be easily isolated from human amnion. In this study, we investigated the therapeutic effect of human amnion-derived MSCs (AMSCs) in rats with severe colitis. Colitis was induced by the administration of 8% dextran sulfate sodium (DSS) from day 0 to day 5, and AMSCs (1 × 106 cells) were transplanted intravenously on day 1. Rats were sacrificed on day 5, and the colon length and histological colitis score were evaluated. The extent of inflammation was evaluated using quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and immunohistochemistry. The effect of AMSCs on the inflammatory signals was investigated in vitro. AMSC transplantation significantly ameliorated the disease activity index score, weight loss, colon shortening, and the histological colitis score. mRNA expression levels of proinflammatory cytokines such as tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and migration inhibitory factor (MIF) were significantly decreased in the rectums of AMSC-treated rats. In addition, the infiltration of monocytes/macrophages was significantly decreased in AMSC-treated rats. In vitro experiments demonstrated that activation of proinflammatory signals induced by TNF-α or lipopolysaccharide (LPS) in immortalized murine macrophage cells (RAW264.7) was significantly attenuated by coculturing with AMSCs or by culturing with a conditioned medium obtained from AMSCs. Although the phosphorylation of IκB induced by TNF-α or LPS was not inhibited by the conditioned medium, nuclear translocation of NF-κB was significantly inhibited by the conditioned medium. Taken together, AMSC transplantation provided significant improvement in rats with severe colitis, possibly through the inhibition of monocyte/macrophage activity and through inhibition of NF-κB activation. AMSCs could be considered as a new cell source for the treatment of severe colitis.
Collapse
Affiliation(s)
- Reizo Onishi
- Department of Gastroenterology and Hepatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Shunsuke Ohnishi
- Department of Gastroenterology and Hepatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Ryosuke Higashi
- Laboratory of Pathophysiology and Therapeutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Michiko Watari
- Department of Gynecology, Tenshi Hospital, Sapporo, Japan
| | - Kenichi Yamahara
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Naoto Okubo
- Laboratory of Pathophysiology and Therapeutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Koji Nakagawa
- Laboratory of Pathophysiology and Therapeutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Takehiko Katsurada
- Department of Gastroenterology and Hepatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Goki Suda
- Department of Gastroenterology and Hepatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Mitsuteru Natsuizaka
- Department of Gastroenterology and Hepatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Hiroshi Takeda
- Laboratory of Pathophysiology and Therapeutics, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Naoya Sakamoto
- Department of Gastroenterology and Hepatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
695
|
Caplan AI. MSCs: The Sentinel and Safe-Guards of Injury. J Cell Physiol 2015; 231:1413-6. [PMID: 26565391 DOI: 10.1002/jcp.25255] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 11/11/2015] [Indexed: 01/08/2023]
Abstract
Mesenchymal stem cells (MSCs) were originally named because they could differentiate in a variety of mesenchymal phenotypes in culture. Evidence indicates that MSCs arise from perivascular cells, pericytes, when the blood vessels are broken or inflamed. These pericyte/MSCs are situated on every blood vessel in the body. The MSCs sense the micro-environment of the injury site and secrete site-specific factors that serve several important reparative functions: first, a curtain of molecules from the front of the MSCs provide a barrier from the interrogation of the over-aggressive immune system. Second, from the back of the MSCs, a different set of bioactive agents inhibit scar formation and establish a regenerative micro-environment. Third, if bacteria are sensed by the MSCs, they produce powerful protein antibiotics that kill the bacteria on contact. Last, the MSCs surround and encyst intruding solid objects like a piece of wood (a "splinter") or other foreign objects. The MSCs act as a combination paramedic and emergency room (ER) staff to survey the damage, isolate foreign components, stabilize the injured tissues, provide antibiotics and encysting protection before a slower, medicinal sequence can be initiated to regenerate the damaged tissue. The MSCs, thus, act as sentinels to safeguard the individual from intrusion and chronic injury. A societal treatment system has evolved, paramedics and ER procedures, which mirror in a macro-sense what MSCs orchestrate in a micro-sense. Key to this new understanding is that MSCs are not "stem cells," but rather as Medicinal Signaling Cells as the therapeutic agents.
Collapse
Affiliation(s)
- Arnold I Caplan
- Department of Biology, Skeletal Research Center, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
696
|
Panchalingam KM, Jung S, Rosenberg L, Behie LA. Bioprocessing strategies for the large-scale production of human mesenchymal stem cells: a review. Stem Cell Res Ther 2015; 6:225. [PMID: 26597928 PMCID: PMC4657237 DOI: 10.1186/s13287-015-0228-5] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Human mesenchymal stem cells (hMSCs), also called mesenchymal stromal cells, have been of great interest in regenerative medicine applications because of not only their differentiation potential but also their ability to secrete bioactive factors that can modulate the immune system and promote tissue repair. This potential has initiated many early-phase clinical studies for the treatment of various diseases, disorders, and injuries by using either hMSCs themselves or their secreted products. Currently, hMSCs for clinical use are generated through conventional static adherent cultures in the presence of fetal bovine serum or human-sourced supplements. However, these methods suffer from variable culture conditions (i.e., ill-defined medium components and heterogeneous culture environment) and thus are not ideal procedures to meet the expected future demand of quality-assured hMSCs for human therapeutic use. Optimizing a bioprocess to generate hMSCs or their secreted products (or both) promises to improve the efficacy as well as safety of this stem cell therapy. In this review, current media and methods for hMSC culture are outlined and bioprocess development strategies discussed.
Collapse
Affiliation(s)
- Krishna M Panchalingam
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
| | - Sunghoon Jung
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada
| | - Lawrence Rosenberg
- Department of Surgery, McGill University Health Centre, 845 Rue Sherbrooke Quest, Montreal, QC, H3G 1A4, Canada.,Jewish General Hospital, 3755 Chemin de la Côte-Ste-Catherine Road, Montreal, QC, H3T 1E2, Canada
| | - Leo A Behie
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, 2500 University Drive NW, Calgary, AB, T2N 1N4, Canada.
| |
Collapse
|
697
|
Effects on Proliferation and Differentiation of Human Umbilical Cord-Derived Mesenchymal Stem Cells Engineered to Express Neurotrophic Factors. Stem Cells Int 2015; 2016:1801340. [PMID: 26649046 PMCID: PMC4663010 DOI: 10.1155/2016/1801340] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 03/29/2015] [Accepted: 04/06/2015] [Indexed: 12/19/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotential cells with capability to form colonies in vitro and differentiate into distinctive end-stage cell types. Although MSCs secrete many cytokines, the efficacy can be improved through combination with neurotrophic factors (NTFs). Moreover, MSCs are excellent opportunities for local delivery of NTFs into injured tissues. The aim of this present study is to evaluate the effects of overexpressing NTFs on proliferation and differentiation of human umbilical cord-derived mesenchymal stem cells (HUMSCs). Overexpressing NTFs had no effect on cell proliferation. Overexpressing NT-3, BDNF, and NGF also had no significant effect on the differentiation of HUMSCs. Overexpressing NTFs all promoted the neurite outgrowth of embryonic chick E9 dorsal root ganglion (DRG). The gene expression profiles of the control and NT-3- and BDNF-modified HUMSCs were compared using RNA sequencing and biological processes and activities were revealed. This study provides novel information about the effects of overexpressing NTFs on HUMSCs and insight into the choice of optimal NTFs for combined cell and gene therapy.
Collapse
|
698
|
Shao J, Zhang W, Yang T. Using mesenchymal stem cells as a therapy for bone regeneration and repairing. Biol Res 2015; 48:62. [PMID: 26530042 PMCID: PMC4630918 DOI: 10.1186/s40659-015-0053-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 10/22/2015] [Indexed: 02/07/2023] Open
Abstract
Bone is a unique tissue which could regenerate completely after injury rather than heal itself with a scar. Compared with other tissues the difference is that, during bone repairing and regeneration, after the inflammatory phase the mesenchymal stem cells (MSCs) are recruited to the injury site and differentiate into either chondroblasts or osteoblasts precursors, leading to bone repairing and regeneration. Besides these two precursors, the MSCs can also differentiate into adipocyte precursors, skeletal muscle precursors and some other mesodermal cells. With this multilineage potentiality, the MSCs are probably used to cure bone injury and other woundings in the near future. Here we will introduce the recent developments in understanding the mechanism of MSCs action in bone regeneration and repairing.
Collapse
Affiliation(s)
- Jin Shao
- Department of Orthopaedics, Shanghai Pudong New Area Gongli Hospital, Second Military Medical University, Shanghai, 200135, China.
| | - Weiwei Zhang
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Tieyi Yang
- Department of Orthopaedics, Shanghai Pudong New Area Gongli Hospital, Second Military Medical University, Shanghai, 200135, China.
| |
Collapse
|
699
|
Ono M, Ohnishi S, Honda M, Ishikawa M, Hosono H, Onishi R, Nakagawa K, Takeda H, Sakamoto N. Effects of human amnion–derived mesenchymal stromal cell transplantation in rats with radiation proctitis. Cytotherapy 2015; 17:1545-59. [DOI: 10.1016/j.jcyt.2015.07.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 06/09/2015] [Accepted: 07/04/2015] [Indexed: 01/25/2023]
|
700
|
Chen YB, Lan YW, Chen LG, Huang TT, Choo KB, Cheng WTK, Lee HS, Chong KY. Mesenchymal stem cell-based HSP70 promoter-driven VEGFA induction by resveratrol alleviates elastase-induced emphysema in a mouse model. Cell Stress Chaperones 2015; 20:979-89. [PMID: 26243699 PMCID: PMC4595438 DOI: 10.1007/s12192-015-0627-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 07/01/2015] [Accepted: 07/19/2015] [Indexed: 01/03/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a sustained blockage of the airways due to lung inflammation occurring with chronic bronchitis and/or emphysema. Progression of emphysema may be slowed by vascular endothelial growth factor A (VEGFA), which reduces apoptotic tissue depletion. Previously, authors of the present report demonstrated that cis-resveratrol (c-RSV)-induced heat-shock protein 70 (HSP70) promoter-regulated VEGFA expression promoted neovascularization of genetically modified mesenchymal stem cells (HSP-VEGFA-MSC) in a mouse model of ischemic disease. Here, this same stem cell line was evaluated for its protective capacity to alleviate elastase-induced pulmonary emphysema in mice. Results of this study showed that c-RSV-treatment of HSP-VEGFA-MSC exhibited synergy between HSP70 transcription activity and induced expression of anti-oxidant-related genes when challenged by cigarette smoke extracts. Eight weeks after jugular vein injection of HSP-VEGFA-MSC into mice with elastase-induced pulmonary emphysema followed by c-RSV treatment to induce transgene expression, significant improvement was observed in respiratory functions. Expression of VEGFA, endogenous nuclear factor erythroid 2-related factor (Nrf 2), and manganese superoxide dismutase (MnSOD) was significantly increased in the lung tissues of the c-RSV-treated mice. Histopathologic examination of treated mice revealed gradual but significant abatement of emphysema and restoration of airspace volume. In conclusion, the present investigation demonstrates that c-RSV-regulated VEGFA expression in HSP-VEGFA-MSC significantly improved the therapeutic effects on the treatment of COPD in the mouse, possibly avoiding side effects associated with constitutive VEGFA expression.
Collapse
Affiliation(s)
- Young-Bin Chen
- Institute of Biotechnology, College of Bioresources and Agriculture, National Taiwan University, Taipei, Taiwan, Republic of China
| | - Ying-Wei Lan
- Graduate Institute of Biomedical Sciences, Division of Biotechnology, Chang Gung University, Tao-Yuan, Taiwan, Republic of China
| | - Lih-Geeng Chen
- Department of Microbiology, Immunology and Biopharmaceuticals, College of Life Sciences, National Chiayi University, Chiayi, 600, Taiwan, Republic of China
| | - Tsung-Teng Huang
- Center for Molecular and Clinical Immunology, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan, Republic of China
| | - Kong-Bung Choo
- Department of Preclinical Sciences, Faculty of Medicine and Health Sciences and Centre for Stem Cell Research, Universiti Tunku Abdul Rahman, Selangor, Malaysia
| | - Winston T K Cheng
- Department of Animal Science and Biotechnology, Tunghai University, Taichung, Taiwan, Republic of China
| | - Hsuan-Shu Lee
- Institute of Biotechnology, College of Bioresources and Agriculture, National Taiwan University, Taipei, Taiwan, Republic of China.
- Department of Internal Medicine, National Taiwan University Hospital, National Taiwan University College of Medicne, Taipei, Taiwan, Republic of China.
| | - Kowit-Yu Chong
- Graduate Institute of Biomedical Sciences, Division of Biotechnology, Chang Gung University, Tao-Yuan, Taiwan, Republic of China.
- Molecular Medicine Research Center, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan, Republic of China.
- Department of Medical Biotechnology and Laboratory Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan, Republic of China.
- Department of Family Medicine, Chang Gung Memorial Hospital-Linkou, Taoyuan, Taiwan, Republic of China.
| |
Collapse
|