Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Tissue Res ; 362(3): 623-32, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26202892

ABSTRACT

In most pre-clinical animal studies investigating stem cell therapy in acute myocardial infarction (AMI), the administered stem cells are isolated from healthy donors. In clinical practice, however, patients who suffer from AMI will receive autologous cells, for example using adipose-derived stem cells (ASC). During AMI, inflammation is induced and we hypothesized that this might affect characteristics of ASC. To investigate this, ASC were isolated from rat adipose tissue 1 day (1D group, n = 5) or 7 days (7D group, n = 6) post-AMI, and were compared with ASC from healthy control rats (Control group, n = 6) and sham-operated rats (Sham 1D group, n = 5). We found that significantly fewer ASC were present 1 day post-AMI in the stromal vascular fraction (SVF), determined by a colony-forming-unit assay (p < 0.001 vs. Control and 7D). These data were confirmed by flow cytometry, showing fewer CD90-positive cells in SVF of the 1D group. When cultured, no differences were found in proliferation rate and cell size between the groups in the first three passages. Also, no difference in the differentiation capacity of ASC was found. In conclusion, it was shown that significantly fewer stem cells were present in the SVF 1 day post-AMI; however, the stem cells that were present showed no functional differences.


Subject(s)
Adipose Tissue/cytology , Myocardial Infarction/pathology , Stem Cells/cytology , Animals , Cell Count , Cell Differentiation , Cell Lineage , Cells, Cultured , Male , Rats, Wistar , Stromal Cells/cytology
2.
Biotechnol Bioeng ; 112(1): 220-7, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25088405

ABSTRACT

The use of stem cells for the repair of damaged cardiac tissue after a myocardial infarction holds great promise. However, a common finding in experimental studies is the low number of cells delivered at the area at risk. To improve the delivery, we are currently investigating a novel delivery platform in which stem cells are conjugated with targeted microbubbles, creating echogenic complexes dubbed StemBells. These StemBells vibrate in response to incoming ultrasound waves making them susceptible to acoustic radiation force. The acoustic force can then be employed to propel circulating StemBells from the centerline of the vessel to the wall, facilitating localized stem cell delivery. In this study, we investigate the feasibility of manipulating StemBells acoustically in vivo after injection using a chicken embryo model. Bare stem cells or unsaturated stem cells (<5 bubbles/cell) do not respond to ultrasound application (1 MHz, peak negative acoustical pressure P_ = 200 kPa, 10% duty cycle). However, stem cells which are fully saturated with targeted microbubbles (>30 bubbles/cell) can be propelled toward and arrested at the vessel wall. The mean translational velocities measured are 61 and 177 µm/s for P- = 200 and 450 kPa, respectively. This technique therefore offers potential for enhanced and well-controlled stem cell delivery for improved cardiac repair after a myocardial infarction.


Subject(s)
Mesenchymal Stem Cell Transplantation , Microbubbles , Microscopy/methods , Stem Cells/cytology , Acoustics , Animals , Cells, Cultured , Chick Embryo , Chickens , Humans
3.
Stem Cell Res ; 13(3 Pt A): 367-78, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25290189

ABSTRACT

Adipose tissue-derived stromal cells (ASC) form a rich source of autologous cells for use in regenerative medicine. In vitro induction of an endothelial phenotype may improve performance of ASCs in cardiovascular repair. Here, we report on an in vitro strategy using direct reprogramming of ASCs by means of ectopic expression of the endothelial-specific transcription factor SRY (sex determining region Y)-box18 (SOX18). SOX18 induces ASCs to express a set of genes involved in vascular patterning: MMP7, KDR, EFNB2, SEMA3G and CXCR4. Accordingly, SOX18 transduced ASCs reorganize under conditions of shear stress, display VEGF-induced chemotaxis and form tubular structures in 3D matrices in an MMP7-dependent manner. These in vitro findings provide insight into molecular and cellular processes downstream of SOX18 and show that reprogramming using SOX18 is sufficient to induce several endothelial-like features in ASCs.


Subject(s)
Adipose Tissue/cytology , Endothelial Cells/metabolism , SOXF Transcription Factors/metabolism , Stromal Cells/metabolism , Cell Differentiation , Cell Movement/drug effects , Cells, Cultured , Cellular Reprogramming , Chemotaxis/drug effects , Endothelial Cells/cytology , Genetic Vectors/genetics , Genetic Vectors/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Matrix Metalloproteinase 7/metabolism , Microtubules/chemistry , Microtubules/metabolism , SOXF Transcription Factors/genetics , Shear Strength , Stromal Cells/cytology , Vascular Endothelial Growth Factor A/pharmacology
4.
Stem Cell Rev Rep ; 10(3): 389-98, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24577790

ABSTRACT

The majority of patients survive an acute myocardial infarction (AMI). Their outcome is negatively influenced by post-AMI events, such as loss of viable cardiomyocytes due to a post-AMI inflammatory response, eventually resulting in heart failure and/or death. Recent pre-clinical animal studies indicate that mesenchymal stem cells derived from adipose tissue (ASC) are new promising candidates that may facilitate cardiovascular regeneration in the infarcted myocardium. In this review we have compared all animal studies in which ASC were used as a therapy post-AMI and have focused on aspects that might be important for future successful clinical application of ASC.


Subject(s)
Mesenchymal Stem Cell Transplantation/methods , Myocardial Infarction/therapy , Adipose Tissue/cytology , Animals , Cells, Cultured , Disease Models, Animal , Humans , Mesenchymal Stem Cells/physiology
5.
Res Vet Sci ; 96(2): 377-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24445251

ABSTRACT

The Wistar rat is a commonly used strain for experimental animal models. Recently it was shown that results vary between studies using Wistar rats of different suppliers. Therefore we studied whether Wistar rats obtained from Harlan Laboratories (Ha, n=24) and Charles River (CR, n=22) had a different outcome in an acute myocardial infarction (AMI) model. AMI was induced in both Ha and CR Wistar rats by one operator. This resulted in a significantly higher survival rate for Ha (79.2±10.2%) compared with CR rats (54.2±10.2%, p<0.05). Furthermore, CR rats had lost significantly more weight after 7 days (-5.9±3.1%) compared with Ha rats (-0.8±1.7%; p<0.001), indicating a worse health status of the CR rats. Paradoxically, the induced infarct was smaller in CR rats (7.3±3.6% of the heart) compared with Ha rats (12.1±4.7%, p<0.05). This indicates that CR rats were less sensitive for the cardiomyocyte damage subsequent to AMI induction, but remarkably showed more clinical side effects indicating that Wistar rats from two suppliers had a different response within the same AMI model.


Subject(s)
Myocardial Infarction/veterinary , Myocytes, Cardiac/ultrastructure , Rats, Wistar/surgery , Animals , Disease Models, Animal , Histocytochemistry , Kaplan-Meier Estimate , Male , Myocardial Infarction/physiopathology , Rats
6.
Minerva Cardioangiol ; 61(6): 617-25, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24253455

ABSTRACT

Inadequate healing following acute myocardial infarction (AMI) can lead to the development of heart failure. The ischemic myocardium triggers an inflammatory response that clears cell debris and initiates the onset of scar tissue formation. The duration and intensity of this inflammatory response have been linked to the cardiac functioning post-AMI. In order to diminish scar tissue formation and stimulate regeneration of cardiac tissue, mesenchymal stem cell (MSC) have been applied post-AMI and showed beneficial effects on cardiac function. However, other than the expected regeneration of cardiac tissue, modulation of the inflammatory response post-AMI, especially related to an effect on monocytes/macrophages, was recently found to be an important aspect of MSC therapy. In healing post-AMI, monocytes and macrophages are key players that can either stimulate or repress inflammation using different phenotypes. Increased levels of the proinflammatory phenotype have clinically been associated with poor cardiac functional outcome post-AMI. MSC have been suggested to switch the monocytes/macrophages phenotype into a more anti-inflammatory state and might therefore beneficially influence the duration and intensity of the inflammatory response and subsequent cardiac function post-AMI. To gain more insight into this effect of MSC, this review provides an overview of the most relevant studies regarding this modulatory effect of MSC on monocytes/macrophages including its mechanisms to improve cardiac functioning post-AMI.


Subject(s)
Inflammation/therapy , Mesenchymal Stem Cell Transplantation/methods , Myocardial Infarction/therapy , Animals , Cicatrix , Heart Failure/etiology , Heart Failure/prevention & control , Humans , Inflammation/physiopathology , Macrophages/metabolism , Mesenchymal Stem Cells/metabolism , Monocytes/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Phenotype , Time Factors
7.
Cell Biol Toxicol ; 28(5): 303-15, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22801743

ABSTRACT

Adipose tissue-derived stem cells (ASCs) are promising candidates for regenerative therapy, like after myocardial infarction. However, when transplanted into the infarcted heart, ASCs are jeopardized by the ischemic environment. Interestingly, it has been shown that multidrug resistance (MDR) proteins like the breast cancer resistance protein (BCRP) and P-glycoprotein (P-gp) have a protective effect in haematopoietic stem cells. In ASC, however, only expression of BCRP was shown until now. In this study, we therefore analysed the expression and functional activity of BCRP and P-gp and their putative function in ischemia in ASC. BCRP and P-gp protein expression was studied over time (passages 2-6) using western blot analysis and immunohistochemical staining. MDR activity was analysed using protein-specific substrate extrusion assays. Ischemia was induced using metabolic inhibition. All analyses demonstrated protein expression and activity of BCRP in ASCs. In contrast, only minor expression of P-gp was found, without functional activity. BCRP expression was most prominent in early passage ASCs (p2) and decreased during culture. Finally, ischemia induced expression of BCRP. In addition, when BCRP was blocked, a significant increase in dead ASCs was found already after 1 h of ischemia. In conclusion, ASCs expressed BCRP, especially in early passages. In addition, we now show for the first time that BCRP protects ASCs against ischemia-induced cell death. These data therefore indicate that for transplantation of ASCs in an ischemic environment, like myocardial infarction, the optimal stem cell protective effect of BCRP theoretically will be achieved with early culture passages ASCs.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Adipose Tissue/metabolism , Gene Expression , Neoplasm Proteins/metabolism , Stem Cells/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Adipose Tissue/cytology , Adult , Biological Transport/genetics , Cell Differentiation , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation , Cell Survival/genetics , Cells, Cultured , Drug Resistance, Neoplasm , Female , Humans , Middle Aged , Models, Biological , Neoplasm Proteins/genetics , Stem Cells/cytology
8.
Cell Tissue Res ; 348(1): 119-30, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22395775

ABSTRACT

Adipose-derived stromal cells (ASC) are promising candidates for cell therapy, for example to treat myocardial infarction. Commonly, fetal bovine serum (FBS) is used in ASC culturing. However, FBS has several disadvantages. Its effects differ between batches and, when applied clinically, transmission of pathogens and antibody development against FBS are possible. In this study, we investigated whether FBS can be substituted by human platelet lysate (PL) in ASC culture, without affecting functional capacities particularly important for cardiac repair application of ASC. We found that PL-cultured ASC had a significant 3-fold increased proliferation rate and a significantly higher attachment to tissue culture plastic as well as to endothelial cells compared with FBS-cultured ASC. PL-cultured ASC remained a significant 25% smaller than FBS-cultured ASC. Both showed a comparable surface marker profile, with the exception of significantly higher levels of CD73, CD90, and CD166 on PL-cultured ASC. PL-cultured ASC showed a significantly higher migration rate compared with FBS-cultured ASC in a transwell assay. Finally, FBS- and PL-cultured ASC had a similar high capacity to differentiate towards cardiomyocytes. In conclusion, this study showed that culturing ASC is more favorable in PL-supplemented medium compared with FBS-supplemented medium.


Subject(s)
Adipose Tissue/cytology , Blood Platelets/metabolism , Blood Substitutes/pharmacology , Cell Extracts/pharmacology , Myocardium/pathology , Serum/metabolism , Wound Healing/drug effects , Adult , Aged , Animals , Biomarkers/metabolism , Blood Platelets/drug effects , Cattle , Cell Adhesion/drug effects , Cell Culture Techniques , Cell Differentiation/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Size/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Flow Cytometry , Humans , Middle Aged , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Stromal Cells/cytology , Stromal Cells/drug effects , Stromal Cells/metabolism
9.
Stem Cell Res ; 7(3): 219-29, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21907165

ABSTRACT

Stem cell therapy is a promising tool to improve outcome after acute myocardial infarction (AMI), but needs to be optimized since results from clinical applications remain ambiguous. A potent source of stem cells is the stromal vascular fraction of adipose tissue (SVF), which contains high numbers of adipose derived stem cells (ASC). We hypothesized that: 1) intravenous injection can be used to apply stem cells to the heart. 2) Uncultured SVF cells are easier and safer when cultured ASCs. 3) Transplantation after the acute inflammation period of AMI is favorable over early injection. For this, AMI was induced in rats by 40min of coronary occlusion. One or seven days after AMI, rats were intravenously injected with vehicle, 5×10(6) uncultured rat SVF cells or 1×10(6) rat ASCs. Rats were analyzed 35 days after AMI. Intravenous delivery of both fresh SVF cells and cultured ASCs 7 days after AMI significantly reduced infarct size compared to vehicle. Similar numbers of stem cells were found in the heart, after treatment with fresh SVF cells and cultured ASCs. Importantly, no adverse effects were found after injection of SVF cells. Using cultured ASCs, however, 3 animals had shortness of breath, and one animal died during injection. In contrast to application at 7 days post AMI, injection of SVF cells 1 day post AMI resulted in a small but non-significant infarct reduction (p=0.35). Taken together, intravenous injection of uncultured SVF cells subsequent to the acute inflammation period, is a promising stem cell therapy for AMI.


Subject(s)
Adipose Tissue/cytology , Myocardial Infarction/pathology , Myocardial Infarction/therapy , Stem Cell Transplantation , Stem Cells/cytology , Animals , Biomarkers/metabolism , Blood Vessels/pathology , Cell Count , Cell Differentiation , Cells, Cultured , Disease Models, Animal , Heart Function Tests , Injections, Intravenous , Macrophages/pathology , Male , Myocardial Infarction/physiopathology , Myocytes, Cardiac/pathology , Rats , Rats, Wistar , Stem Cell Transplantation/adverse effects , Stromal Cells/cytology , Stromal Cells/transplantation , Thromboembolism/etiology , Thromboembolism/pathology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...