Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
EBioMedicine ; 60: 102989, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32920368

ABSTRACT

BACKGROUND: Stem cells` (SC) functional heterogeneity and its poorly understood aetiology impedes clinical development of cell-based therapies in regenerative medicine and oncology. Recent studies suggest a strong correlation between the SC migration potential and their therapeutic efficacy in humans. Designating SC migration as a denominator of functional SC heterogeneity, we sought to identify highly migrating subpopulations within different SC classes and evaluate their therapeutic properties in comparison to the parental non-selected cells. METHODS: We selected highly migrating subpopulations from mesenchymal and neural SC (sMSC and sNSC), characterized their features including but not limited to migratory potential, trophic factor release and transcriptomic signature. To assess lesion-targeted migration and therapeutic properties of isolated subpopulations in vivo, surgical transplantation and intranasal administration of MSCs in mouse models of glioblastoma and Alzheimer's disease respectively were performed. FINDINGS: Comparison of parental non-selected cells with isolated subpopulations revealed superior motility and migratory potential of sMSC and sNSC in vitro. We identified podoplanin as a major regulator of migratory features of sMSC/sNSC. Podoplanin engineering improved oncovirolytic activity of virus-loaded NSC on distantly located glioblastoma cells. Finally, sMSC displayed more targeted migration to the tumour site in a mouse glioblastoma model and remarkably higher potency to reduce pathological hallmarks and memory deficits in transgenic Alzheimer's disease mice. INTERPRETATION: Functional heterogeneity of SC is associated with their motility and migration potential which can serve as predictors of SC therapeutic efficacy. FUNDING: This work was supported in part by the Robert Bosch Stiftung (Stuttgart, Germany) and by the IZEPHA grant.


Subject(s)
Cell Movement , Stem Cells/physiology , Alzheimer Disease/therapy , Animals , Biomarkers , Cell Survival , Cell Tracking/methods , Cells, Cultured , Disease Models, Animal , Gene Expression , Gene Expression Profiling , Humans , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Mice , Mice, Transgenic , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Oncolytic Virotherapy , Stem Cell Transplantation , Stem Cells/cytology , Treatment Outcome
2.
EBioMedicine ; 60: 102987, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32942121

ABSTRACT

BACKGROUND: Limited knowledge of stem cell therapies` mechanisms of action hampers their sustainable implementation into the clinic. Specifically, the interactions of transplanted stem cells with the host vasculature and its implications for their therapeutic efficacy are not elucidated. We tested whether adhesion receptors and chemokine receptors on stem cells can be functionally modulated, and consequently if such modulation may substantially affect therapeutically relevant stem cell interactions with the host endothelium. METHODS: We investigated the effects of cationic molecule polyethylenimine (PEI) treatment with or without nanoparticles on the functions of adhesion receptors and chemokine receptors of human bone marrow-derived Mesenchymal Stem Cells (MSC). Analyses included MSC functions in vitro, as well as homing and therapeutic efficacy in rodent models of central nervous system´s pathologies in vivo. FINDINGS: PEI treatment did not affect viability, immunomodulation or differentiation potential of MSC, but increased the CCR4 expression and functionally blocked their adhesion receptors, thus decreasing their adhesion capacity in vitro. Intravenously applied in a rat model of brain injury, the homing rate of PEI-MSC in the brain was highly increased with decreased numbers of adherent PEI-MSC in the lung vasculature. Moreover, in comparison to untreated MSC, PEI-MSC featured increased tumour directed migration in a mouse glioblastoma model, and superior therapeutic efficacy in a murine model of stroke. INTERPRETATION: Balanced stem cell adhesion and migration in different parts of the vasculature and tissues together with the local microenvironment impacts their therapeutic efficacy. FUNDING: Robert Bosch Stiftung, IZEPHA grant, EU grant 7 FP Health.


Subject(s)
Cell Adhesion , Cell Movement , Endothelium/metabolism , Stem Cells/metabolism , Animals , Biomarkers , Cell Differentiation , Cell Line , Cell- and Tissue-Based Therapy , Cells, Cultured , Cellular Microenvironment , Disease Models, Animal , Glioma/diagnosis , Glioma/pathology , Glioma/therapy , Humans , Immunophenotyping , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Mice , Rats , Stem Cell Transplantation , Xenograft Model Antitumor Assays
3.
Transfusion ; 58(5): 1132-1142, 2018 05.
Article in English | MEDLINE | ID: mdl-29473177

ABSTRACT

BACKGROUND: Endothelial colony-forming progenitor cells (ECFCs) are promising candidates for cell therapies. However, ECFC translation to the clinic requires optimized isolation and manufacture technologies according to good manufacturing practice (GMP). STUDY DESIGN AND METHODS: ECFCs were manufactured from steady-state peripheral blood (PB) leukapheresis (11 donors), using GMP-compliant technologies including pooled human platelet (PLT) lysate, and compared to human umbilical cord endothelial cells, human aortic endothelial cells, and human cerebral microvascular endothelial cells. Specific variables assessed were growth kinetics, phenotype, trophic factors production, stimulation of tube formation, and Dil-AcLDL uptake. RESULTS: ECFCs could be isolated from PB leukapheresis units with mean processed volume of 5411 mL and mean white blood cell (WBC) concentration factor of 8.74. The mean frequency was 1.44 × 10-8 ECFCs per WBC, corresponding to a mean of 177.8 ECFCs per apheresis unit. Expandable for up to 12 cumulative population doublings, calculated projection showed that approximately 730 × 103 ECFCs could be manufactured from 1 apheresis unit. ECFCs produced epidermal growth factor, hepatocyte growth factor, vascular endothelial growth factor (VEGF)-A, PLT-derived growth factor-B, interleukin-8, and monocyte chemoattractant protein-1, featured high potential for capillary-like tubes formation, and showed no telomerase activity. They were characterized by CD29, CD31, CD44, CD105, CD117, CD133, CD144, CD146, and VEGF-R2 expression, with the most common subpopulation CD34+CD117-CD133-. Compared to controls, ECFCs featured greater Dil-AcLDL uptake and higher expression of CD29, CD31, CD34, CD44, CD144, and VEGF-R2. CONCLUSIONS: Here we show that isolation of ECFCs with proangiogenic profile from steady-state PB leukapheresis is feasible, marking a first step toward ECFC product manufacture according to GMP.


Subject(s)
Blood Platelets/chemistry , Endothelial Progenitor Cells/cytology , Leukapheresis , Peripheral Blood Stem Cells/cytology , Antigens, CD/analysis , Cell-Free System/chemistry , Humans , Lipoproteins, LDL/analysis , Manufactured Materials
4.
Cell Transplant ; 23 Suppl 1: S123-39, 2014.
Article in English | MEDLINE | ID: mdl-25302802

ABSTRACT

In view of the rapid preclinical development of cell-based therapies for neurodegenerative disorders, traumatic brain injury, and tumors, the safe and efficient delivery and targeting of therapeutic cells to the central nervous system is critical for maintaining therapeutic efficacy and safety in the respective disease models. Our previous data demonstrated therapeutically efficacious and targeted delivery of mesenchymal stem cells (MSCs) to the brain in the rat 6-hydroxydopamine model of Parkinson's disease (PD). The present study examined delivery of bone marrow-derived MSCs, macrophages, and microglia to the brain in a transgenic model of PD [(Thy1)-h[A30P] αS] and an APP/PS1 model of Alzheimer's disease (AD) via intranasal application (INA). INA of microglia in naive BL/6 mice led to targeted and effective delivery of cells to the brain. Quantitative PCR analysis of eGFP DNA showed that the brain contained the highest amount of eGFP-microglia (up to 2.1 × 10(4)) after INA of 1 × 10(6) cells, while the total amount of cells detected in peripheral organs did not exceed 3.4 × 10(3). Seven days after INA, MSCs expressing eGFP were detected in the olfactory bulb (OB), cortex, amygdala, striatum, hippocampus, cerebellum, and brainstem of (Thy1)-h[A30P] αS transgenic mice, showing predominant distribution within the OB and brainstem. INA of eGFP-expressing macrophages in 13-month-old APP/PS1 mice led to delivery of cells to the OB, hippocampus, cortex, and cerebellum. Both MSCs and macrophages contained Iba-1-positive population of small microglia-like cells and Iba-1-negative large rounded cells showing either intracellular amyloid ß (macrophages in APP/PS1 model) or α-synuclein [MSCs in (Thy1)-h[A30P] αS model] immunoreactivity. Here, we show, for the first time, intranasal delivery of cells to the brain of transgenic PD and AD mouse models. Additional work is needed to determine the optimal dosage (single treatment regimen or repeated administrations) to achieve functional improvement in these mouse models with intranasal microglia/macrophages and MSCs. This manuscript is published as part of the International Association of Neurorestoratology (IANR) special issue of Cell Transplantation.


Subject(s)
Alzheimer Disease/therapy , Brain/pathology , Macrophages/transplantation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Microglia/transplantation , Parkinson Disease/therapy , Administration, Intranasal , Animals , Biomarkers/metabolism , Bone Marrow Cells/cytology , Cell Differentiation , Cell Lineage , Disease Models, Animal , Female , Flow Cytometry , Green Fluorescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
5.
Stem Cells Dev ; 23(10): 1097-108, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24405418

ABSTRACT

Bone marrow stromal cells (BMSCs) are composed of progenitor and multipotent skeletal stem cells, which are able to differentiate in vitro into osteocytes, adipocytes, and chondrocytes. Mouse BMSCs (mBMSCs) are a versatile model system to investigate factors involved in BMSC differentiation in vitro and in vivo as a variety of transgenic mouse models are available. In this study, mBMSCs were isolated and osteogenic differentiation was investigated in tissue culture and in vivo. Three out of seven independent cell isolates showed the ability to differentiate into osteocytes, adipocytes, and chondrocytes in vitro. In vitro multipotency of an established mBMSC line was maintained over 45 passages. The osteogenic differentiation of this cell line was confirmed by quantitative polymerase chain reaction (qPCR) analysis of specific markers such as osteocalcin and shown to be Runx2 dependent. Notably, the cell line, when transplanted subcutaneously into mice, possesses full skeletal stem cell characteristics in vivo in early and late passages, evident from bone tissue formation, induction of vascularization, and hematopoiesis. This cell line provides, thus, a versatile tool to unravel the molecular mechanisms governing osteogenesis in vivo thereby aiding to improve current strategies in bone regenerative therapy.


Subject(s)
Bone Marrow Cells/metabolism , Cell Differentiation , Multipotent Stem Cells/metabolism , Osteogenesis , Animals , Bone Marrow Cells/cytology , Core Binding Factor Alpha 1 Subunit/metabolism , Mice , Multipotent Stem Cells/cytology , Stromal Cells/cytology , Stromal Cells/metabolism
6.
BMC Med ; 11: 146, 2013 Jun 11.
Article in English | MEDLINE | ID: mdl-23758701

ABSTRACT

BACKGROUND: Mesenchymal stromal cells (MSCs) are attractive for cell-based therapies ranging from regenerative medicine and tissue engineering to immunomodulation. However, clinical efficacy is variable and it is unclear how the phenotypes defining bone marrow (BM)-derived MSCs as well as donor characteristics affect their functional properties. METHODS: BM-MSCs were isolated from 53 (25 female, 28 male; age: 13 to 80 years) donors and analyzed by: (1) phenotype using flow cytometry and cell size measurement; (2) in vitro growth kinetics using population doubling time; (3) colony formation capacity and telomerase activity; and (4) function by in vitro differentiation capacity, suppression of T cell proliferation, cytokines and trophic factors secretion, and hormone and growth factor receptor expression. Additionally, expression of Oct4, Nanog, Prdm14 and SOX2 mRNA was compared to pluripotent stem cells. RESULTS: BM-MSCs from younger donors showed increased expression of MCAM, VCAM-1, ALCAM, PDGFRß, PDL-1, Thy1 and CD71, and led to lower IL-6 production when co-cultured with activated T cells. Female BM-MSCs showed increased expression of IFN-γR1 and IL-6ß, and were more potent in T cell proliferation suppression. High-clonogenic BM-MSCs were smaller, divided more rapidly and were more frequent in BM-MSC preparations from younger female donors. CD10, ß1integrin, HCAM, CD71, VCAM-1, IFN-γR1, MCAM, ALCAM, LNGFR and HLA ABC were correlated to BM-MSC preparations with high clonogenic potential and expression of IFN-γR1, MCAM and HLA ABC was associated with rapid growth of BM-MSCs. The mesodermal differentiation capacity of BM-MSCs was unaffected by donor age or gender but was affected by phenotype (CD10, IFN-γR1, GD2). BM-MSCs from female and male donors expressed androgen receptor and FGFR3, and secreted VEGF-A, HGF, LIF, Angiopoietin-1, basic fibroblast growth factor (bFGF) and NGFB. HGF secretion correlated negatively to the expression of CD71, CD140b and Galectin 1. The expression of Oct4, Nanog and Prdm14 mRNA in BM-MSCs was much lower compared to pluripotent stem cells and was not related to donor age or gender. Prdm14 mRNA expression correlated positively to the clonogenic potential of BM-MSCs. CONCLUSIONS: By identifying donor-related effects and assigning phenotypes of BM-MSC preparations to functional properties, we provide useful tools for assay development and production for clinical applications of BM-MSC preparations.


Subject(s)
Immunophenotyping , Living Donors , Mesenchymal Stem Cells/immunology , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Cell Differentiation/immunology , Cells, Cultured , Female , Humans , Immunophenotyping/methods , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/pathology , Middle Aged , Sex Factors , Young Adult
7.
Cytotherapy ; 15(3): 292-306, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23312449

ABSTRACT

BACKGROUND AIMS: Mesenchymal stromal/stem cells (MSCs) can be isolated from human bone marrow (BM), expanded ex vivo and identified via numerous surface antigens. Despite the importance of these cells in regenerative therapy programs, it is unclear whether the cell membrane signature defining MSC preparations ex vivo is determined during culture or may reflect an in vivo counterpart. BM-MSC phenotype in vivo requires further investigation. METHODS: To characterize cells in their natural BM environment, we performed multi-parametric immunohistochemistry on trabecular bone biopsy specimens from multiple donors and described cells by different morphology and micro-anatomic localization in relationship to a precise pattern of MSC antigen expression. RESULTS: Microscopically examined high-power field marrow sections revealed an overlapping in vivo expression of antigens characterizing ex vivo expanded BM-MSCs, including CD10, CD73, CD140b, CD146, GD2 and CD271. Expanding this panel to proteins associated with pluripotency, such as Oct4, Nanog and SSEA-4, we were able to identify different cellular populations in the human trabecular bone and BM expressing different progenitor cell markers. CONCLUSIONS: Targeting several multipotency and pluripotency markers, we found that the BM contains identifiable and distinct progenitor cells further justifying their introduction for a wide range of applications in regenerative medicine.


Subject(s)
Antigens, CD/metabolism , Cell Differentiation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Biomarkers/metabolism , Bone Marrow/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Gene Expression , Homeodomain Proteins/metabolism , Humans , Mesenchymal Stem Cells/cytology , Nanog Homeobox Protein , Octamer Transcription Factor-3/metabolism , Stage-Specific Embryonic Antigens/metabolism
8.
Stem Cells Dev ; 21(13): 2457-70, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22309203

ABSTRACT

Despite their paracrine activites, cardiomyogenic differentiation of bone marrow (BM)-derived mesenchymal stem cells (MSCs) is thought to contribute to cardiac regeneration. To systematically evaluate the role of differentiation in MSC-mediated cardiac regeneration, the cardiomyogenic differentiation potential of human MSCs (hMSCs) and murine MSCs (mMSCs) was investigated in vitro and in vivo by inducing cardiomyogenic and noncardiomyogenic differentiation. Untreated hMSCs showed upregulation of cardiac tropopin I, cardiac actin, and myosin light chain mRNA and protein, and treatment of hMSCs with various cardiomyogenic differentiation media led to an enhanced expression of cardiomyogenic genes and proteins; however, no functional cardiomyogenic differentiation of hMSCs was observed. Moreover, co-culturing of hMSCs with cardiomyocytes derived from murine pluripotent cells (mcP19) or with murine fetal cardiomyocytes (mfCMCs) did not result in functional cardiomyogenic differentiation of hMSCs. Despite direct contact to beating mfCMCs, hMSCs could be effectively differentiated into cells of only the adipogenic and osteogenic lineage. After intramyocardial transplantation into a mouse model of myocardial infarction, Sca-1(+) mMSCs migrated to the infarcted area and survived at least 14 days but showed inconsistent evidence of functional cardiomyogenic differentiation. Neither in vitro treatment nor intramyocardial transplantation of MSCs reliably generated MSC-derived cardiomyocytes, indicating that functional cardiomyogenic differentiation of BM-derived MSCs is a rare event and, therefore, may not be the main contributor to cardiac regeneration.


Subject(s)
Bone Marrow/metabolism , Cell Differentiation , Mesenchymal Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Actins/metabolism , Adult , Animals , Antigens, CD/metabolism , Cell Lineage , Cell Movement , Coculture Techniques/methods , Culture Media/metabolism , Female , Flow Cytometry , Gene Expression Regulation , Humans , Male , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred C57BL , Middle Aged , Models, Animal , Myocardial Infarction/metabolism , Myocardial Infarction/therapy , Myocytes, Cardiac/cytology , Myosin Light Chains/metabolism , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Troponin I/metabolism , Tumor Cells, Cultured
9.
Br J Haematol ; 153(4): 520-8, 2011 May.
Article in English | MEDLINE | ID: mdl-21418181

ABSTRACT

Incompatible blood group antigens are highly immunogenic and can cause graft rejections. Focusing on distinct carbohydrate- and protein-based membrane structures, defined by blood group antigens, we investigated human bone marrow-derived mesenchymal stem cells (MSCs) cultured in human serum. The presence of H (CD173), ABO, RhD, RhCE, RhAG, Kell, urea transporter type B (SLC14A1, previously known as JK), and Duffy antigen receptor of chemokines (DARC) was evaluated at the levels of genome, transcriptome and antigen. Fucosyltransferase-1 (FUT1), RHCE, KEL, SLC14A1 (JK) and DARC mRNA were transcribed in MSCs. FUT1 mRNA transcription was lost during differentiation. The mRNA transcription of SLC14A1 (JK) decreased during chondrogenic differentiation, while that of DARC increased during adipogenic differentiation. All MSCs synthesized SLC14A1 (JK) but no DARC protein. However, none of the protein antigens tested occurred on the surface, indicating a lack of associated protein function in the membrane. As A and B antigens are neither expressed nor adsorbed, concerns of ABO compatibility with human serum supplements during culture are alleviated. The H antigen expression by GD2dim+ MSCs identified two distinct MSC subpopulations and enabled their isolation. We hypothesize that GD2(dim+) H(+) MSCs retain a better 'stemness'. Because immunogenic blood group antigens are lacking, they cannot affect MSC engraftment in vivo, which is promising for clinical applications.


Subject(s)
Blood Group Antigens/metabolism , Mesenchymal Stem Cells/metabolism , ABO Blood-Group System/metabolism , Blood Group Antigens/genetics , Cell Differentiation/genetics , Cells, Cultured , Duffy Blood-Group System/biosynthesis , Duffy Blood-Group System/genetics , Erythrocytes/metabolism , Gangliosides/metabolism , Humans , Immunophenotyping , Membrane Transport Proteins/biosynthesis , Membrane Transport Proteins/genetics , Mesenchymal Stem Cells/cytology , RNA, Messenger/genetics , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Transcription, Genetic , Urea Transporters
10.
BMC Cell Biol ; 11: 22, 2010 Apr 06.
Article in English | MEDLINE | ID: mdl-20370915

ABSTRACT

BACKGROUND: For clinical applications of mesenchymal stem cells (MSCs), labeling and tracking is crucial to evaluate cell distribution and homing. Magnetic resonance imaging (MRI) has been successfully established detecting MSCs labeled with superparamagnetic particles of iron oxide (SPIO). Despite initial reports that labeling of MSCs with SPIO is safe without affecting the MSC's biology, recent studies report on influences of SPIO-labeling on metabolism and function of MSCs. Exposition of cells and tissues to high magnetic fields is the functional principle of MRI. In this study we established innovative labeling protocols for human MSCs using clinically established SPIO in combination with magnetic fields and investigated on functional effects (migration assays, quantification of colony forming units, analyses of gene and protein expression and analyses on the proliferation capacity, the viability and the differentiation potential) of magnetic fields on unlabeled and labeled human MSCs. To evaluate the imaging properties, quantification of the total iron load per cell (TIL), electron microscopy, and MRI at 3.0 T were performed. RESULTS: Human MSCs labeled with SPIO permanently exposed to magnetic fields arranged and grew according to the magnetic flux lines. Exposure of MSCs to magnetic fields after labeling with SPIO significantly enhanced the TIL compared to SPIO labeled MSCs without exposure to magnetic fields resulting in optimized imaging properties (detection limit: 1,000 MSCs). Concerning the TIL and the imaging properties, immediate exposition to magnetic fields after labeling was superior to exposition after 24 h. On functional level, exposition to magnetic fields inhibited the ability of colony formation of labeled MSCs and led to an enhanced expression of lipoprotein lipase and peroxisome proliferator-activated receptor-gamma in labeled MSCs under adipogenic differentiation, and to a reduced expression of alkaline phosphatase in unlabeled MSCs under osteogenic differentiation as detected by qRT-PCR. Moreover, microarray analyses revealed that exposition of labeled MSCs to magnetic fields led to an up regulation of CD93 mRNA and cadherin 7 mRNA and to a down regulation of Zinc finger FYVE domain mRNA. Exposition of unlabeled MSCs to magnetic fields led to an up regulation of CD93 mRNA, lipocalin 6 mRNA, sialic acid acetylesterase mRNA, and olfactory receptor mRNA and to a down regulation of ubiquilin 1 mRNA. No influence of the exposition to magnetic fields could be observed on the migration capacity, the viability, the proliferation rate and the chondrogenic differentiation capacity of labeled or unlabeled MSCs. CONCLUSIONS: In our study an innovative labeling protocol for tracking MSCs by MRI using SPIO in combination with magnetic fields was established. Both, SPIO and the static magnetic field were identified as independent factors which affect the functional biology of human MSCs. Further in vivo investigations are needed to elucidate the molecular mechanisms of the interaction of magnetic fields with stem cell biology.


Subject(s)
Ferrosoferric Oxide , Magnetics , Mesenchymal Stem Cells/cytology , Nanoparticles , Cell Differentiation , Cell Proliferation , Humans , Magnetic Resonance Imaging
11.
Transplantation ; 87(9 Suppl): S45-9, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19424005

ABSTRACT

Mesenchymal stem cells (MSC) are a type of multipotent progenitor cell, originally isolated from the bone marrow. In addition to multilineage differentiation and participation in the hematopoietic niche, they exert powerful immunomodulatory effects, which include inhibition of proliferation and function of T cells, B cells, and natural killer cells. These unique properties make MSC of great interest for clinical applications in tissue engineering and immunosuppression. Underlying the MSC-mediated immunomodulatory mechanisms is a nonspecific antiproliferative effect, which is the consequence of cyclin D2 inhibition. Of special interest are the molecular mechanisms, by which MSC influence their target cells. Several studies have been conducted in this field, and the current data suggest roles for indoleamine 2,3-dioxygenase, prostaglandin E2, nitric oxide, histocompatibility locus antigen-G, insulin-like growth factor-binding proteins, and tolerogenic antigen-presenting cells. Understanding these mechanisms is crucial for future use of MSC in research and clinical applications.


Subject(s)
Mesenchymal Stem Cells/immunology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Bone Marrow Cells/physiology , Cell Division , Dinoprostone/physiology , HLA Antigens/physiology , HLA-G Antigens , Hematopoiesis , Histocompatibility Antigens Class I/physiology , Humans , Immune Tolerance , Immunosuppression Therapy , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Insulin-Like Growth Factor Binding Proteins/physiology , Killer Cells, Natural/immunology , Lymphocyte Activation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Nitric Oxide/physiology , T-Lymphocytes/immunology
12.
Cytotherapy ; 11(1): 68-78, 2009.
Article in English | MEDLINE | ID: mdl-19191056

ABSTRACT

BACKGROUND AIMS: Labeling of stem cells is crucial to allow tracking of stem cell homing and engraftment after transplantation. In this study we evaluated the influence of cell labeling procedures using clinically approved small particles of iron oxide (SPIO) with or without transfection reagents (TA) on functional parameters of human mesenchymal stem cells (MSC). METHODS: The study was approved by the institutional review board of the University of Tubingen, Germany. Seven populations of bone marrow (BM)-derived human mesenchymal stem cells (MSC) were labeled with SPIO alone or in combination with various TA. Directly after labeling and two passages after labeling migration assays, quantification of colony-forming units and quantitative evaluation of the differentiation potential were performed. Quantification of the cellular total iron load (TIL), determination of the cellular viability and electron microscopy were also performed. RESULTS: Labeling of mesenchymal stem cells with SPIO with or without TA did not affect cell viability and differentiation potential significantly. SPIO in combination with TA coated the cellular surface directly after labeling but was incorporated into the cells after two passages. Labeling of mesenchymal stem cells with TA led to a significant decrease of migration capacity. This effect was abolished after two passages. Labeling with and without TA led to a significant decrease in colony formation ability. This effect could also be observed after two passages. CONCLUSIONS: The observed decrease of migration capacity and colony-formation ability was not associated with either TIL or localization of particles of iron oxide. SPIO labeling with and without TA had functional effects on human mesenchymal stem cells by decreasing the migration capacity and colony-formation ability of the stem cells.


Subject(s)
Cell Movement/drug effects , Contrast Media/pharmacology , Iron/pharmacology , Mesenchymal Stem Cells/drug effects , Oxides/pharmacology , Staining and Labeling/methods , Adolescent , Adult , Aged , Bone Marrow Cells/drug effects , Bone Marrow Cells/physiology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Survival/drug effects , Cell Survival/physiology , Dextrans , Female , Ferrosoferric Oxide , Gene Expression/drug effects , Gene Expression/physiology , Humans , Magnetite Nanoparticles , Male , Mesenchymal Stem Cells/physiology , Mesenchymal Stem Cells/ultrastructure , Microscopy, Electron, Transmission , Middle Aged , Young Adult
13.
Cell Transplant ; 17(8): 923-41, 2008.
Article in English | MEDLINE | ID: mdl-19069635

ABSTRACT

Multiple sclerosis (MS) is an inflammatory and demyelinating disease of the central nervous system (CNS). Mesenchymal stem cells (MSC) have been shown to ameliorate symptoms in experimental autoimmune encephalomyelitis (EAE), a model of MS. Using cloned MSC labeled with clinically approved small particles of iron oxide (SPIO) for treatment of EAE we analyzed the tissue localization of transferred cells. Treatment with unlabeled MSC led to disease amelioration compared to controls. In contrast, treatment with SPIO-labeled MSC lead to increase in disease severity. Treatment with SPIO alone did not alter disease course. After transplantation labeled and nonlabeled MSC were detected in the CNS and the liver with significantly more SPIO-labeled cells present in the CNS. Iron deposition was present in the group treated with SPIO-labeled MSC, indicating that in vivo the initially cell surface-bound iron detached from the MSC. These results could be of great importance for imaging of patients in the clinical setting, indicating that in vivo application of SPIO-labeled MSC needs to be performed with caution because the cell-derived exposure of iron can lead to disease aggravation.


Subject(s)
Coloring Agents/adverse effects , Encephalomyelitis, Autoimmune, Experimental/surgery , Ferric Compounds/adverse effects , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/drug effects , Staining and Labeling/methods , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Central Nervous System/cytology , Central Nervous System/pathology , Central Nervous System/physiopathology , Coloring Agents/metabolism , Disease Models, Animal , Disease Progression , Female , Ferric Compounds/metabolism , Graft Survival/drug effects , Graft Survival/physiology , Liver/cytology , Liver/pathology , Liver/physiopathology , Magnetic Resonance Imaging , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Microscopy, Electron, Transmission , Multiple Sclerosis/surgery , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...