Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Cells ; 8(12)2019 11 20.
Article in English | MEDLINE | ID: mdl-31757119

ABSTRACT

BACKGROUND: Ventricular arrhythmias (VA) are a common cause of sudden death after myocardial infarction (MI). Therefore, developing new therapeutic methods for the prevention and treatment of VA is of prime importance. METHODS: Human bone marrow derived CD271+ mesenchymal stem cells (MSC) were tested for their antiarrhythmic effect. This was done through the development of a novel mouse model using an immunocompromised Rag2-/- γc-/- mouse strain subjected to myocardial "infarction-reinfarction". The mice underwent a first ischemia-reperfusion through the left anterior descending (LAD) artery closure for 45 minutes with a subsequent second permanent LAD ligation after seven days from the first infarct. RESULTS: This mouse model induced various types of VA detected with continuous electrocardiogram (ECG) monitoring via implanted telemetry device. The immediate intramyocardial delivery of CD271+ MSC after the first MI significantly reduced VA induced after the second MI. CONCLUSIONS: In addition to the clinical relevance, more closely reflecting patients who suffer from severe ischemic heart disease and related arrhythmias, our new mouse model bearing reinfarction warrants the time required for stem cell engraftment and for the first time enables us to analyze and verify significant antiarrhythmic effects of human CD271+ stem cells in vivo.


Subject(s)
Adapalene/immunology , Anti-Arrhythmia Agents/therapeutic use , Disease Models, Animal , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Myocardial Infarction/therapy , Adapalene/analysis , Animals , Female , Humans , Immunophenotyping , Mice , Mice, Knockout
2.
Cells ; 9(1)2019 Dec 27.
Article in English | MEDLINE | ID: mdl-31892273

ABSTRACT

BACKGROUND: Bone marrow (BM)-derived stem cells with their various functions and characteristics have become a well-recognized source for the cell-based therapies. However, knowledge on their therapeutic potential and the shortage for a cross-link between distinct BM-derived stem cells, primed after the onset of myocardial infarction (MI), seems to be still rudimentary. Therefore, the post-examination of the therapeutic characteristics of such primed hematopoietic CD133+ and mesenchymal CD271+ stem cells was the object of the present study. METHODS AND RESULTS: The effects of respective CD133+ and CD271+ mononuclear cells alone as well as in the co-culture model have been explored with focus on their angiogenic potential. The phenotypic analysis revealed a small percentage of isolated cells expressing both surface markers. Moreover, target stem cells isolated with our standardized immunomagnetic isolation procedure did not show any negative alterations following BM storage in regard to cell numbers and/or quality. In vitro network formation relied predominantly on CD271+ stem cells when compared with single CD133+ culture. Interestingly, CD133+ cells contributed in the tube formation, only if they were cultivated in combination with CD271+ cells. Additional to the in vitro examination, therapeutic effects of the primed stem cells were investigated 48 h post MI in a murine model. Hence, we have found a lower expression of transforming growth factor ßeta 3 (TGFß3) as well as an increase of the proangiogenic factors after CD133+ cell treatment in contrast to CD271+ cell treatment. On the other hand, the CD271+ cell therapy led to a lower expression of the inflammatory cytokines. CONCLUSION: The interactions between CD271+ and CD133+ subpopulations the extent to which the combination may enhance cardiac regeneration has still not been investigated so far. We expect that the multiple characteristics and various regenerative effects of CD271+ cells alone as well as in combination with CD133+ will result in an improved therapeutic impact on ischemic heart disease.


Subject(s)
AC133 Antigen/metabolism , Adapalene/metabolism , Cell Differentiation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Myocardial Infarction/therapy , Neovascularization, Physiologic , Animals , Biomarkers , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Proliferation , Disease Models, Animal , Female , Fluorescent Antibody Technique , Gene Expression Profiling , Immunophenotyping , Mesenchymal Stem Cell Transplantation/methods , Mice , Mice, Transgenic , Myocardial Infarction/etiology , Regeneration
3.
Dis Model Mech ; 11(6)2018 06 22.
Article in English | MEDLINE | ID: mdl-29752300

ABSTRACT

Ischemic heart failure is the leading cause of mortality worldwide. An early boost of intracardiac regenerative key mechanisms and angiogenetic niche signaling in cardiac mesenchymal stem cells (MSCs) could improve myocardial infarction (MI) healing. Epicardial erythropoietin (EPO; 300 U kg-1) was compared with intraperitoneal and intramyocardial EPO treatments after acute MI in rats (n=156). Real-time PCR and confocal microscopy revealed that epicardial EPO treatment enhanced levels of intracardiac regenerative key indicators (SDF-1, CXCR4, CD34, Bcl-2, cyclin D1, Cdc2 and MMP2), induced transforming growth factor ß (TGF-ß)/WNT signaling in intramyocardial MSC niches through the direct activation of AKT and upregulation of upstream signals FOS and Fzd7, and augmented intracardiac mesenchymal proliferation 24 h after MI. Cardiac catheterization and tissue analysis showed superior cardiac functions, beneficial remodeling and increased capillary density 6 weeks after MI. Concomitant fluorescence-activated cell sorting, co-cultures with neonatal cardiomyocytes, angiogenesis assays, ELISA, western blotting and RAMAN spectroscopy demonstrated that EPO could promote cardiomyogenic differentiation that was specific of tissue origin and enhance paracrine angiogenetic activity in cardiac CD45-CD44+DDR2+ MSCs. Epicardial EPO delivery might be the optimal route for efficient upregulation of regenerative key signals after acute MI. Early EPO-mediated stimulation of mesenchymal proliferation, synergistic angiogenesis with cardiac MSCs and direct induction of TGF-ß/WNT signaling in intramyocardial cardiac MSCs could initiate an accelerated healing process that enhances cardiac recovery.


Subject(s)
Erythropoietin/therapeutic use , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Myocardial Ischemia/pathology , Myocardial Ischemia/therapy , Myocardium/pathology , Neovascularization, Physiologic , Pericardium/metabolism , Acute Disease , Animals , Antigens, CD/metabolism , Capillaries/pathology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Erythropoietin/administration & dosage , Erythropoietin/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Heart Function Tests , Heart Ventricles/drug effects , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Hemodynamics/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Mesenchymal Stem Cells/drug effects , Mesoderm/pathology , Myocardial Ischemia/physiopathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Neovascularization, Physiologic/drug effects , Rats , Rats, Inbred Lew , Regeneration/drug effects , Transforming Growth Factor beta/metabolism , Wnt Signaling Pathway/drug effects
4.
Sci Rep ; 7(1): 9755, 2017 08 29.
Article in English | MEDLINE | ID: mdl-28852100

ABSTRACT

Different subtypes of bone marrow-derived stem cells are characterized by varying functionality and activity after transplantation into the infarcted heart. Improvement of stem cell therapeutics requires deep knowledge about the mechanisms that mediate the benefits of stem cell treatment. Here, we demonstrated that co-transplantation of mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs) led to enhanced synergistic effects on cardiac remodeling. While HSCs were associated with blood vessel formation, MSCs were found to possess transdifferentiation capacity. This cardiomyogenic plasticity of MSCs was strongly promoted by a gap junction-dependent crosstalk between myocytes and stem cells. The inhibition of cell-cell coupling significantly reduced the expression of the cardiac specific transcription factors NKX2.5 and GATA4. Interestingly, we observed that small non-coding RNAs are exchanged between MSCs and cardiomyocytes in a GJ-dependent manner that might contribute to the transdifferentiation process of MSCs within a cardiac environment. Our results suggest that the predominant mechanism of HSCs contribution to cardiac regeneration is based on their ability to regulate angiogenesis. In contrast, transplanted MSCs have the capability for intercellular communication with surrounding cardiomyocytes, which triggers the intrinsic program of cardiogenic lineage specification of MSCs by providing cardiomyocyte-derived cues.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/physiology , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/physiology , Myocardial Infarction/therapy , Signal Transduction , Animals , Cell Communication , Cell Transdifferentiation , Cells, Cultured , Disease Models, Animal , Gap Junctions/metabolism , Humans , Mice, SCID , Myocytes, Cardiac/physiology , Neovascularization, Physiologic
5.
Stem Cell Res Ther ; 8(1): 33, 2017 02 10.
Article in English | MEDLINE | ID: mdl-28187777

ABSTRACT

BACKGROUND: CD133+ stem cells represent a promising subpopulation for innovative cell-based therapies in cardiovascular regeneration. Several clinical trials have shown remarkable beneficial effects following their intramyocardial transplantation. Yet, the purification of CD133+ stem cells is typically performed in centralized clean room facilities using semi-automatic manufacturing processes based on magnetic cell sorting (MACS®). However, this requires time-consuming and cost-intensive logistics. METHODS: CD133+ stem cells were purified from patient-derived sternal bone marrow using the recently developed automatic CliniMACS Prodigy® BM-133 System (Prodigy). The entire manufacturing process, as well as the subsequent quality control of the final cell product (CP), were realized on-site and in compliance with EU guidelines for Good Manufacturing Practice. The biological activity of automatically isolated CD133+ cells was evaluated and compared to manually isolated CD133+ cells via functional assays as well as immunofluorescence microscopy. In addition, the regenerative potential of purified stem cells was assessed 3 weeks after transplantation in immunodeficient mice which had been subjected to experimental myocardial infarction. RESULTS: We established for the first time an on-site manufacturing procedure for stem CPs intended for the treatment of ischemic heart diseases using an automatized system. On average, 0.88 × 106 viable CD133+ cells with a mean log10 depletion of 3.23 ± 0.19 of non-target cells were isolated. Furthermore, we demonstrated that these automatically isolated cells bear proliferation and differentiation capacities comparable to manually isolated cells in vitro. Moreover, the automatically generated CP shows equal cardiac regeneration potential in vivo. CONCLUSIONS: Our results indicate that the Prodigy is a powerful system for automatic manufacturing of a CD133+ CP within few hours. Compared to conventional manufacturing processes, future clinical application of this system offers multiple benefits including stable CP quality and on-site purification under reduced clean room requirements. This will allow saving of time, reduced logistics and diminished costs.


Subject(s)
Automation, Laboratory/instrumentation , Cell Separation/instrumentation , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/physiology , Myocardial Infarction/therapy , Regeneration/physiology , AC133 Antigen/genetics , AC133 Antigen/metabolism , Aged , Animals , Biomarkers/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Cell Differentiation , Cell Proliferation , Cell Separation/methods , Disease Models, Animal , Female , Gene Expression , Hematopoietic Stem Cells/cytology , Humans , Male , Mice , Mice, SCID , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Recovery of Function/physiology , Tissue Donors
6.
Stem Cells Int ; 2016: 7152761, 2016.
Article in English | MEDLINE | ID: mdl-27795713

ABSTRACT

Aim. CD133+ stem cells bear huge potential for regenerative medicine. However, low retention in the injured tissue and massive cell death reduce beneficial effects. In order to address these issues, we intended to develop a nonviral system for appropriate cell engineering. Materials and Methods. Modification of human CD133+ stem cells with magnetic polyplexes carrying microRNA was studied in terms of efficiency, safety, and targeting potential. Results. High microRNA uptake rates (~80-90%) were achieved without affecting CD133+ stem cell properties. Modified cells can be magnetically guided. Conclusion. We developed a safe and efficient protocol for CD133+ stem cell modification. Our work may become a basis to improve stem cell therapeutical effects as well as their monitoring with magnetic resonance imaging.

7.
Forsch Komplementmed ; 22(4): 246-50, 2015.
Article in English | MEDLINE | ID: mdl-26565406

ABSTRACT

BACKGROUND: Sauna bathing is claimed to provide benefits for patients suffering from cardiovascular diseases. The current study aims at analyzing the induction of potential regenerative processes by quantifying the mobilization of bone marrow-derived stem cells into the peripheral blood of healthy adults following Finnish sauna. MATERIALS AND METHODS: Twenty healthy unbiased male volunteers (20-30 years old) were exposed to a Finnish sauna bath (3 × 10 min, 90°C). Venous blood samples were drawn before (baseline), immediately, and 6 h as well as 24 h after the sauna bath. Blood analysis included isolation of mononuclear cells, cell staining with mononuclear antibodies, and fluorescence-activated cell sorting (FACS). For baseline and 24 h post-sauna samples colony-forming unit-Hill assays were applied to quantify endothelial progenitor cells (EPC). RESULTS: Flow cytometry revealed an upregulation of circulating CD45+/CD309+ progenitor cells immediately after the sauna bath, however without reaching statistical significance. Circulating cell numbers of the CD45+CD34+, CD45+CD34+CD133+, and CD45+CD34+CD117+ populations did not show clear enhancements following sauna. EPC colony formation tended to be enhanced after sauna as compared to baseline values. CONCLUSION: Peripheral EPC numbers exhibited a moderate increase following Finnish sauna in a cohort of healthy young men. Furthermore, sauna bathing tended to increase EPC colony-forming capacity. These rather weak responses to thermotherapy might indicate a ceiling effect. In individuals exhibiting cardiovascular risk factors the effects may be more pronounced.


Subject(s)
Endothelial Progenitor Cells/cytology , Steam Bath , Adult , Antigens, CD/metabolism , Cell Movement/physiology , Endothelial Progenitor Cells/metabolism , Flow Cytometry , Hemodynamics/physiology , Humans , Male , Young Adult
8.
Cell Physiol Biochem ; 37(1): 77-93, 2015.
Article in English | MEDLINE | ID: mdl-26303045

ABSTRACT

BACKGROUND/AIMS: CD117(+) stem cell (SC) based therapy is considered an alternative therapeutic option for terminal heart disease. However, controversies exist on the effects of CD117(+) SC implantation. In particular, the link between CD117(+) SC function and angiotensin-II-type-2 receptor (AT2R) after MI is continuously discussed. We therefore asked whether 1) AT2R stimulation influences CD117(+) SC properties in vitro and, 2) which effects can be ascribed to AT2R stimulation in vivo. METHODS: We approached AT2R stimulation with Angiotensin II while simultaneously blocking its opponent receptor AT1 with Losartan. CD117 effects were dissected using a 2D-Matrigel assay and HL-1 co-culture in vitro. A model of myocardial infarction, in which we implanted EGFP(+) CD117 SC, was further applied. RESULTS: While we found indications for AT2R driven vasculogenesis in vitro, co-culture experiments revealed that CD117(+) SC improve vitality of cardiomyocytes independently of AT2R function. Likewise, untreated CD117(+) SC had a positive effect on cardiac function and acted cardioprotective in vivo. CONCLUSIONS: Therefore, our data show that transient AT2R stimulation does not significantly add to the beneficial actions of CD117(+) SC in vivo. Yet, exploiting AT2R driven vasculogenis via an optimized AT2R stimulation protocol may become a promising tool for cardiac SC therapy.


Subject(s)
Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Proto-Oncogene Proteins c-kit/metabolism , Receptor, Angiotensin, Type 2/metabolism , Stem Cells/metabolism , Stem Cells/physiology , Angiotensin II/metabolism , Animals , Cell Line , Cell- and Tissue-Based Therapy/methods , Coculture Techniques/methods , Losartan/pharmacology , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Stem Cells/drug effects
9.
J Cell Mol Med ; 19(8): 1975-85, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25991381

ABSTRACT

Myocardial infarction (MI) is a major condition causing heart failure (HF). After MI, the renin angiotensin system (RAS) and its signalling octapeptide angiotensin II (Ang II) interferes with cardiac injury/repair via the AT1 and AT2 receptors (AT1R, AT2R). Our study aimed at deciphering the mechanisms underlying the link between RAS and cellular components of the immune response relying on a rodent model of HF as well as HF patients. Flow cytometric analyses showed an increase in the expression of CD4(+) AT2R(+) cells in the rat heart and spleen post-infarction, but a reduction in the peripheral blood. The latter was also observed in HF patients. The frequency of rat CD4(+) AT2R(+) T cells in circulating blood, post-infarcted heart and spleen represented 3.8 ± 0.4%, 23.2 ± 2.7% and 22.6 ± 2.6% of the CD4(+) cells. CD4(+) AT2R(+) T cells within blood CD4(+) T cells were reduced from 2.6 ± 0.2% in healthy controls to 1.7 ± 0.4% in patients. Moreover, we characterized CD4(+) AT2R(+) T cells which expressed regulatory FoxP3, secreted interleukin-10 and other inflammatory-related cytokines. Furthermore, intramyocardial injection of MI-induced splenic CD4(+) AT2R(+) T cells into recipient rats with MI led to reduced infarct size and improved cardiac performance. We defined CD4(+) AT2R(+) cells as a T cell subset improving heart function post-MI corresponding with reduced infarction size in a rat MI-model. Our results indicate CD4(+) AT2R(+) cells as a promising population for regenerative therapy, via myocardial transplantation, pharmacological AT2R activation or a combination thereof.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Heart Function Tests , Myocardial Infarction/immunology , Myocardial Infarction/physiopathology , Receptor, Angiotensin, Type 2/metabolism , Ventricular Remodeling , Animals , Cardiotonic Agents/metabolism , Heart Failure/blood , Heart Failure/complications , Heart Failure/immunology , Heart Failure/physiopathology , Humans , Immunomodulation , Interleukin-10/blood , Myocardial Infarction/blood , Myocardial Infarction/complications , Myocardial Ischemia/blood , Myocardial Ischemia/complications , Myocardial Ischemia/immunology , Myocardial Ischemia/physiopathology , Rats, Wistar , Tumor Necrosis Factor-alpha/blood
10.
Cell Transplant ; 24(11): 2409-22, 2015.
Article in English | MEDLINE | ID: mdl-24800805

ABSTRACT

Stem cell transplantation is a viable strategy for regenerative medicine. However, it is inevitable to have cells undergo storage for several hours or days due to processing and transportation. Therefore, it is crucial to have rigidly controlled conditions ensuring the therapeutic benefit of isolated stem cells. In the present study, we investigated the impact of short-term storage on human CD133(+) cells. CD133(+) cells were isolated from human bone marrow and kept at standardized nonfreezing storage conditions for up to 72 h. Cell viability (apoptosis/necrosis) and expression of CD133 and CXCR4 were analyzed by flow cytometry. Metabolic activity was determined using an MTT assay; colony-forming ability, as well as endothelial-like differentiation, was further evaluated. A qRT-PCR array was employed to investigate the expression of stemness genes. CD133 and CXCR4 expressions were preserved at all time points. After 30 h, cell number and metabolic activity decreased, although no significant changes were detected in cell viability and proliferation as well as endothelial-like differentiation. Cell viability and proliferation decreased significantly only after 72 h of storage. Our results indicate that storage of isolated human CD133(+) bone marrow stem cells in liquid allows for high viability and functionality. However, storage time should be limited in order to avoid cell loss.


Subject(s)
Cell Survival/physiology , Stem Cells/cytology , Tissue Preservation , AC133 Antigen , Antigens, CD/genetics , Antigens, CD/metabolism , Bone Marrow Cells/cytology , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Peptides/genetics , Peptides/metabolism , Receptors, CXCR4 , Stem Cell Transplantation
11.
Stem Cells Int ; 2014: 197154, 2014.
Article in English | MEDLINE | ID: mdl-24799915

ABSTRACT

Genetic modifications of bone marrow derived human mesenchymal stem cells (hMSCs) using microRNAs (miRs) may be used to improve their therapeutic potential and enable innovative strategies in tissue regeneration. However, most of the studies use cultured hMSCs, although these can lose their stem cell characteristics during expansion. Therefore, we aimed to develop a nonviral miR carrier based on polyethylenimine (PEI) bound to magnetic nanoparticles (MNPs) for efficient miR delivery in freshly isolated hMSCs. MNP based transfection is preferable for genetic modifications in vivo due to improved selectivity, safety of delivery, and reduced side effects. Thus, in this study different miR/PEI and miR/PEI/MNP complex formulations were tested in vitro for uptake efficiency and cytotoxicity with respect to the influence of an external magnetic field. Afterwards, optimized magnetic complexes were selected and compared to commercially available magnetic vectors (Magnetofectamine, CombiMag). We found that all tested transfection reagents had high miR uptake rates (yielded over 60%) and no significant cytotoxic effects. Our work may become crucial for virus-free introduction of therapeutic miRs as well as other nucleic acids in vivo. Moreover, in the field of targeted stem cell therapy nucleic acid delivery prior to transplantation may allowfor initial cell modulation in vitro.

12.
Bone Marrow Res ; 2014: 182645, 2014.
Article in English | MEDLINE | ID: mdl-25610653

ABSTRACT

Both stem cell chemokine stromal cell-derived factor-1α (SDF-1α) and extracellular nucleotides such as adenosine triphosphate (ATP) are increased in ischemic myocardium. Since ATP has been reported to influence cell migration, we analysed the migratory response of bone marrow cells towards a combination of SDF-1 and ATP. Total nucleated cells (BM-TNCs) were isolated from bone marrow of cardiac surgery patients. Migration assays were performed in vitro. Subsequently, migrated cells were subjected to multicolor flow cytometric analysis of CD133, CD34, CD117, CD184, CD309, and CD14 expression. BM-TNCs migrated significantly towards a combination of SDF-1 and ATP. The proportions of CD34+ cells as well as subpopulations coexpressing multiple stem cell markers were selectively enhanced after migration towards SDF-1 or SDF-1 + ATP. After spontaneous migration, significantly fewer stem cells and CD184+ cells were detected. Direct incubation with SDF-1 led to a reduction of CD184+ but not stem cell marker-positive cells, while incubation with ATP significantly increased CD14+ percentage. In summary, we found that while a combination of SDF-1 and ATP elicited strong migration of BM-TNCs in vitro, only SDF-1 was responsible for selective attraction of hematopoietic stem cells. Meanwhile, spontaneous migration of stem cells was lower compared to BM-TNCs or monocytes.

13.
Nanomedicine (Lond) ; 9(7): 999-1017, 2014 May.
Article in English | MEDLINE | ID: mdl-24063366

ABSTRACT

AIM: Magnetically guided transfection has been shown as a promising approach for the genetic modification of cells. We observed that polyethylenimine (PEI)-condensed pDNA, combined with magnetic nanoparticles (MNPs) via biotin-streptavidin interactions could provide higher transfection efficiency than pDNA/PEI alone, even without the application of a magnetic force. Therefore, we intended to investigate the beneficial properties of MNP-based transfection. MATERIALS & METHODS: We performed three-color fluorescent labeling of magnetic transfection complexes and traced them inside human mesenchymal stem cells over time using confocal microscopy in order to study pDNA release kinetics by colocalization studies. RESULTS: We demonstrated that MNP-combined pDNA/PEI complexes provide more rapid and efficient release of pDNA than pDNA/PEI alone, which could be explained by the retention of PEI on the surface of the MNPs due to strong biotin-streptavidin interactions. CONCLUSION: The process of pDNA liberation may significantly influence the efficiency of the transfection vector. Therefore, it should be carefully considered when creating novel gene delivery agents.


Subject(s)
Mesenchymal Stem Cells/metabolism , Plasmids/chemistry , Transfection/methods , Biotin/chemistry , Humans , Plasmids/genetics , Polyethyleneimine/chemistry , Streptavidin/chemistry
14.
Stem Cells Int ; 2013: 698076, 2013.
Article in English | MEDLINE | ID: mdl-24194767

ABSTRACT

Human Mesenchymal Stem Cells (hMSCs) present a promising tool for regenerative medicine. However, ex vivo expansion is necessary to obtain sufficient cells for clinical therapy. Conventional growth media usually contain the critical component fetal bovine serum. For clinical use, chemically defined media will be required. In this study, the capability of two commercial, chemically defined, serum-free hMSC growth media (MSCGM-CD and PowerStem) for hMSC proliferation was examined and compared to serum-containing medium (MSCGM). Immunophenotyping of hMSCs was performed using flow cytometry, and they were tested for their ability to differentiate into a variety of cell types. Although the morphology of hMSCs cultured in the different media differed, immunophenotyping displayed similar marker patterns (high expression of CD29, CD44, CD73, and CD90 cell surface markers and absence of CD45). Interestingly, the expression of CD105 was significantly lower for hMSCs cultured in MSCGM-CD compared to MSCGM. Both groups maintained mesenchymal multilineage differentiation potential. In conclusion, the serum-free growth medium is suitable for hMSC culture and comparable to its serum-containing counterpart. As the expression of CD105 has been shown to positively influence hMSC cardiac regenerative potential, the impact of CD105 expression onto clinical use after expansion in MSCGM-CD will have to be tested.

15.
Int J Mol Sci ; 14(6): 10710-26, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23702843

ABSTRACT

Bone marrow derived human mesenchymal stem cells (hMSCs) show promising potential in regeneration of defective tissue. Recently, gene silencing strategies using microRNAs (miR) emerged with the aim to expand the therapeutic potential of hMSCs. However, researchers are still searching for effective miR delivery methods for clinical applications. Therefore, we aimed to develop a technique to efficiently deliver miR into hMSCs with the help of a magnetic non-viral vector based on cationic polymer polyethylenimine (PEI) bound to iron oxide magnetic nanoparticles (MNP). We tested different magnetic complex compositions and determined uptake efficiency and cytotoxicity by flow cytometry. Additionally, we monitored the release, processing and functionality of delivered miR-335 with confocal laser scanning microscopy, real-time PCR and live cell imaging, respectively. On this basis, we established parameters for construction of magnetic non-viral vectors with optimized uptake efficiency (~75%) and moderate cytotoxicity in hMSCs. Furthermore, we observed a better transfection performance of magnetic complexes compared to PEI complexes 72 h after transfection. We conclude that MNP-mediated transfection provides a long term effect beneficial for successful genetic modification of stem cells. Hence, our findings may become of great importance for future in vivo applications.


Subject(s)
Gene Transfer Techniques , Magnetite Nanoparticles/chemistry , Mesenchymal Stem Cells/metabolism , MicroRNAs/metabolism , Cell Movement , Gene Knockdown Techniques , Humans , Mesenchymal Stem Cells/cytology , MicroRNAs/genetics , Time Factors , Transfection
16.
J Cell Mol Med ; 17(6): 766-73, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23560719

ABSTRACT

Periodontitis is a bacterially induced chronic inflammatory disease. Dental follicle progenitor cells (DFPCs) have been proposed as biological graft for periodontal regenerative therapies. The potential impact of bacterial toxins on DFPCs properties is still poorly understood. The aim of this study was to investigate whether DFPCs are able to sense and respond to lipopolysaccharide (LPS) from Porphyromonas gingivalis, a major periopathogenic bacterium. Specifically, we hypothesized that LPS could influence the migratory capacity and IL-6 secretion of DFPCs. DFPCs properties were compared to bone marrow mesenchymal stem cells (BMSCs), a well-studied class of adult stem cells. The analysis by flow cytometry indicated that DFPCs, similar to BMSCs, expressed low levels of both toll-like receptor (TLR) 2 and 4. The TLR4 mRNA expression was down-regulated in response to LPS in both cell populations, while on protein level TLR4 was significantly up-regulated on BMSCs. The TLR2 expression was not influenced by the LPS treatment in both DFPCs and BMSCs. The migratory efficacy of LPS-treated DFPCs was evaluated by in vitro scratch wound assays and found to be significantly increased. Furthermore, we assayed the secretion of interleukin-6 (IL-6), a potent stimulator of cell migration. Interestingly, the levels of IL-6 secretion of DFPCs and BMSCs remained unchanged after the LPS treatment. Taken together, these results suggest that DFPCs are able to sense and respond to P. gingivalis LPS. Our study provides new insights into understanding the physiological role of dental-derived progenitor cells in sites of periodontal infection.


Subject(s)
Dental Sac/drug effects , Lipopolysaccharides/pharmacology , Mesenchymal Stem Cells/drug effects , Porphyromonas gingivalis/chemistry , Adolescent , Antigens, CD/genetics , Antigens, CD/metabolism , Biomarkers/metabolism , Cell Movement/drug effects , Colony-Forming Units Assay , Dental Sac/cytology , Dental Sac/metabolism , Gene Expression , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Primary Cell Culture , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Young Adult
17.
Cell Transplant ; 22(9): 1627-35, 2013.
Article in English | MEDLINE | ID: mdl-23051098

ABSTRACT

Human bone marrow stem cell populations have been applied for cardiac regeneration purposes within different clinical settings in the recent past. The migratory capacity of applied stem cell populations towards injured tissue, after undergoing specific peri-interventional harvesting and isolation procedures, represents a key factor limiting therapeutic efficacy. We therefore aimed at analyzing the migratory capacity of human cluster of differentiation (CD) 133(+) bone marrow stem cells in vivo after intraoperative harvesting from the sternal bone marrow. Human CD133(+) bone marrow stem cells were isolated from the sternal bone marrow of patients undergoing cardiac surgery at our institution. Migratory capacity towards stromal cell-derived factor-1α (SDF-1α) gradients was tested in vitro and in vivo by intravital fluoresecence microscopy, utilizing the cremaster muscle model in severe combined immunodeficient (SCID) mice and analyzing CD133(+) cell interaction with the local endothelium. Furthermore, the role of a local inflammatory stimulus for CD133(+) cell interaction with the endothelium was studied. In order to describe endothelial response upon chemokine stimulation laser scanning microscopy of histological cremaster muscle samples was performed. SDF-1α alone was capable to induce relevant early CD133(+) cell interaction with the endothelium, indicated by the percentage of rolling CD133(+) cells (45.9±1.8% in "SDF-1" vs. 17.7±2.7% in "control," p<0.001) and the significantly reduced rolling velocity after SDF-1α treatment. Furthermore, SDF-1α induced firm endothelial adhesion of CD133(+) cells in vivo. Firm endothelial adhesion, however, was significantly enhanced by additional inflammatory stimulation with tumor necrosis factor-α (TNF-α) (27.9±4.3 cells/mm(2)in "SDF-1 + TNF" vs. 2.2±1.1 cells/mm(2) in "control," p<0.001). CD133(+) bone marrow stem cells exhibit sufficient in vivo homing towards SDF-1α gradients in an inflammatory microenvironment after undergoing standardized intraoperative harvesting and isolation from the sternal bone marrow.


Subject(s)
Antigens, CD/metabolism , Bone Marrow Cells/cytology , Cell Movement/physiology , Glycoproteins/metabolism , Peptides/metabolism , Stem Cells/cytology , AC133 Antigen , Animals , Bone Marrow Cells/metabolism , Cell Differentiation/physiology , Cell Separation/methods , Humans , Male , Mice , Mice, SCID , Microscopy, Confocal , Receptors, CXCR4/metabolism , Stem Cells/metabolism , Tumor Necrosis Factor-alpha/metabolism
18.
PLoS One ; 7(7): e39490, 2012.
Article in English | MEDLINE | ID: mdl-22844395

ABSTRACT

This study assessed the concept of whether delivery of magnetic nanobeads (MNBs)/adenoviral vectors (Ad)-encoded hVEGF gene (Ad(hVEGF)) could regenerate ischaemically damaged hearts in a rat acute myocardial infarction model under the control of an external magnetic field. Adenoviral vectors were conjugated to MNBs with the Sulfo-NHS-LC-Biotin linker. In vitro transduction efficacy of MNBs/Ad-encoded luciferase gene (Ad(luc)) was compared with Ad(luc) alone in human umbilical vein endothelial cells (HUVECs) under magnetic field stimulation. In vivo, in a rat acute myocardial infarction (AMI) model, MNBs/Ad(hVEGF) complexes were injected intravenously and an epicardial magnet was employed to attract the circulating MNBs/Ad(hVEGF) complexes. In vitro, compared with Ad(luc) alone, MNBs/Ad(luc) complexes had a 50-fold higher transduction efficiency under the magnetic field. In vivo, epicardial magnet effectively attracted MNBs/Ad(hVEGF) complexes and resulted in strong therapeutic gene expression in the ischemic zone of the infarcted heart. When compared to other MI-treated groups, the MI-M(+)/Ad(hVEGF) group significantly improved left ventricular function (p<0.05) assessed by pressure-volume loops after 4 weeks. Also the MI-M(+)/Ad(hVEGF) group exhibited higher capillary and arteriole density and lower collagen deposition than other MI-treated groups (p<0.05). Magnetic targeting enhances transduction efficiency and improves heart function. This novel method to improve gene therapy outcomes in AMI treatment offers the potential into clinical applications.


Subject(s)
Adenoviridae/genetics , Heart/physiopathology , Magnets , Nanoparticles , Regeneration , Transfection/methods , Vascular Endothelial Growth Factor A/genetics , Animals , Arterioles/metabolism , Capillaries/metabolism , Feasibility Studies , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Magnetic Fields , Male , Myocardial Infarction/genetics , Myocardial Infarction/physiopathology , Myocardial Infarction/therapy , Nanoparticles/adverse effects , Rats
19.
J Cell Mol Med ; 15(9): 1989-98, 2011 Sep.
Article in English | MEDLINE | ID: mdl-20629995

ABSTRACT

Transplantation of mesenchymal stem cells (MSCs) derived from adult bone marrow has been proposed as a potential therapeutic approach for post-infarction left ventricular (LV) dysfunction. However, age-related functional decline of stem cells has restricted their clinical benefits after transplantation into the infarcted myocardium. The limitations imposed on patient cells could be addressed by genetic modification of stem cells. This study was designed to improve our understanding of genetic modification of human bone marrow derived mesenchymal stem cells (hMSCs) by polyethylenimine (PEI, branched with Mw 25 kD), one of non-viral vectors that show promise in stem cell genetic modification, in the context of cardiac regeneration for patients. We optimized the PEI-mediated reporter gene transfection into hMSCs, evaluated whether transfection efficiency is associated with gender or age of the cell donors, analysed the influence of cell cycle on transfection and investigated the transfer of therapeutic vascular endothelial growth factor gene (VEGF). hMSCs were isolated from patients with cardiovascular disease aged from 41 to 85 years. Optimization of gene delivery to hMSCs was carried out based on the particle size of the PEI/DNA complexes, N/P ratio of complexes, DNA dosage and cell viability. The highest efficiency with the cell viability near 60% was achieved at N/P ratio 2 and 6.0 µg DNA/cm(2) . The average transfection efficiency for all tested samples, middle-age group (<65 years), old-age group (>65 years), female group and male group was 4.32%, 3.85%, 4.52%, 4.14% and 4.38%, respectively. The transfection efficiency did not show any correlation either with the age or the gender of the donors. Statistically, there were two subpopulations in the donors; and transfection efficiency in each subpopulation was linearly related to the cell percentage in S phase. No significant phenotypic differences were observed between these two subpopulations. Furthermore, PEI-mediated therapeutic gene VEGF transfer could significantly enhance the expression level.


Subject(s)
Bone Marrow Cells/metabolism , Gene Transfer Techniques , Mesenchymal Stem Cells/metabolism , Polyethyleneimine/pharmacology , Adult , Aged , Aged, 80 and over , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Cell Death/drug effects , Cell Survival/drug effects , DNA/metabolism , Female , Green Fluorescent Proteins/metabolism , Humans , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Middle Aged , Phenotype , S Phase/drug effects , Transfection , Vascular Endothelial Growth Factor A/metabolism
20.
Atherosclerosis ; 207(1): 116-22, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19423111

ABSTRACT

OBJECTIVE: The bulk of LDL entrapped in the arterial intima is modified by hydrolytic enzymes, leading to extensive cleavage of cholesterylesters and liberation of fatty acids. The latter induce apoptosis in endothelial cells but are far less cytotoxic towards macrophages. We have compared the cytotoxic effects of enzymatically modified LDL (E-LDL) on macrophages and polymorphonuclear granulocytes (PMN). METHODS AND RESULTS: E-LDL displayed toxicity towards PMN at far lower concentrations than towards monocyte-derived macrophages. Native or oxidized LDL had no effect. Free fatty acids contained in E-LDL were the cause of the observed toxicity, which could be mimicked by linoleic acid, oleic acid and arachidonic acid. E-LDL provoked Ca(2+) influx and activated PMN, as witnessed by the generation of superoxide anions and peroxidase secretion. Inhibition of either oxidative burst or calcium influx did not diminish the cytotoxicity of E-LDL. Similar to free linoleic acid, E-LDL lysed red blood cells and rapidly rendered cells permeable to propidium iodide. CONCLUSION: Possibly through their capacity to directly perturb cell membranes, free fatty acids contained in E-LDL exert potent cytotoxic effects on PMN. This may be one reason why PMN are not abundantly present in atherosclerotic lesions, and why PMN-depletion suppresses atherogenesis.


Subject(s)
Atherosclerosis/metabolism , Fatty Acids, Nonesterified/metabolism , Lipoproteins, LDL/metabolism , Macrophages/metabolism , Neutrophils/metabolism , Peptide Hydrolases/metabolism , Sterol Esterase/metabolism , Adenosine Triphosphate/metabolism , Animals , Arachidonic Acid/metabolism , Atherosclerosis/pathology , Calcium/metabolism , Cell Death , Cell Membrane Permeability , Cell Survival , Cells, Cultured , Hemolysis , Humans , Hydrolysis , L-Lactate Dehydrogenase/metabolism , Linoleic Acid/metabolism , Macrophages/pathology , Neutrophils/pathology , Oleic Acid/metabolism , Peroxidase/metabolism , Rabbits , Respiratory Burst , Superoxides/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...