Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cardiovasc Res ; 108(1): 39-49, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26187727

ABSTRACT

RATIONALE: Engraftment and survival of transplanted stem or stromal cells in the microenvironment of host tissues may be improved by combining such cells with scaffolds to delay apoptosis and enhance regenerative properties. AIMS: We examined whether poly(lactic-co-glycolic acid) pharmacologically active microcarriers (PAMs) releasing vascular endothelial growth factor (VEGF) enhance survival, differentiation, and angiogenesis of adipose tissue-mesenchymal stromal cells (AT-MSCs). We analysed the efficacy of transplanted AT-MSCs conjugated with PAMs in a murine model of acute myocardial infarction (AMI). METHODS AND RESULTS: We used fibronectin-coated (empty) PAMs or VEGF-releasing PAMs covered with murine AT-MSCs. Twelve-month-old C57 mice underwent coronary artery ligation to induce AMI, and were randomized into five treatment groups: AMI control (saline 20 µL, n = 7), AMI followed by intramyocardial injection with AT-MSCs (2.5 × 10(5) cells/20 µL, n = 5), or concentrated medium (CM) from AT-MSCs (20 µL, n = 8), or AT-MSCs (2.5 × 10(5) cells/20 µL) conjugated with empty PAMs (n = 7), or VEGF-releasing PAMs (n = 8). Sham-operated mice (n = 7) were used as controls. VEGF-releasing PAMs increased proliferation and angiogenic potential of AT-MSCs, but did not impact their osteogenic or adipogenic differentiation. AT-MSCs conjugated with VEGF-releasing PAMs inhibited apoptosis, decreased fibrosis, increased arteriogenesis and the number of cardiac-resident Ki-67 positive cells, and improved myocardial fractional shortening compared with AT-MSCs alone when transplanted into the infarcted hearts of C57 mice. With the exception of fractional shortening, all such effects of AT-MSCs conjugated with VEGF-PAMs were paralleled by the injection of CM. CONCLUSIONS: AT-MSCs conjugated with VEGF-releasing PAMs exert paracrine effects that may have therapeutic applications.


Subject(s)
Adipose Tissue/cytology , Mesenchymal Stem Cell Transplantation , Myocardial Infarction/therapy , Vascular Endothelial Growth Factor A/metabolism , Animals , Cells, Cultured , Male , Mice , Mice, Inbred C57BL , Microspheres
2.
J Biomed Mater Res A ; 103(9): 3012-25, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25727843

ABSTRACT

One of the main cause of ineffective cell therapy in repairing the damaged heart is the poor yield of grafted cells. To overcome this drawback, rats with 4-week-old myocardial infarction (MI) were injected in the border zone with human adipose-derived stem cells (ADSCs) conveyed by poly(lactic-co-glycolic acid) microcarriers (PAMs) releasing hepatocyte growth factor (HGF) and insulin-like growth factor-1 (IGF-1) (GFsPAMs). According to treatments, animals were subdivided into different groups: MI_ADSC, MI_ADSC/PAM, MI_GFsPAM, MI_ADSC/GFsPAM, and untreated MI_V. Two weeks after injection, a 31% increase in ADSC engraftment was observed in MI_ADSC/PAM compared with MI_ADSC (p < 0.05). A further ADSC retention was obtained in MI_ADSC/GFsPAM with respect to MI_ADSC (106%, p < 0.05) and MI_ADSC/PAM (57%, p < 0.05). A 130% higher density of blood vessels of medium size was present in MI_ADSC/GFsPAM compared with MI_ADSC (p < 0.01). MI_ADSC/GFsPAM also improved, albeit slightly, left ventricular remodeling and hemodynamics with respect to the other groups. Notably, ADSCs and/or PAMs, with or without HGF/IGF-1, trended to induce arrhythmias in electrically driven, Langendorff-perfused, hearts of all groups. Thus, PAMs releasing HGF/IGF-1 markedly increase ADSC engraftment 2 weeks after injection and stimulate healing in chronically infarcted myocardium, but attention should be paid to potentially negative electrophysiological consequences.


Subject(s)
Hepatocyte Growth Factor/administration & dosage , Insulin-Like Growth Factor I/administration & dosage , Myocardial Infarction/drug therapy , Myocardial Infarction/therapy , Stem Cell Transplantation/methods , Adipose Tissue/cytology , Animals , Arrhythmias, Cardiac/etiology , Biomimetic Materials/chemistry , Disease Models, Animal , Drug Carriers/administration & dosage , Humans , Lactic Acid , Male , Materials Testing , Microspheres , Myocardial Infarction/pathology , Neovascularization, Physiologic/drug effects , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Rats , Rats, Wistar , Stem Cell Transplantation/adverse effects , Ventricular Remodeling , Wound Healing/drug effects
3.
J Biomed Mater Res A ; 103(5): 1828-39, 2015 May.
Article in English | MEDLINE | ID: mdl-25098676

ABSTRACT

For tissue-engineering studies of the infarcted heart it is essential to identify a source of cells that may provide cardiomyocyte progenitors, which is easy to amplify, accessible in adults, and allowing autologous grafts. Preclinical studies have shown that human adipose-derived stem cells (ADSCs) can differentiate into cardiomyocyte-like cells and improve heart function in myocardial infarction. We have developed pharmacologically active microcarriers (PAMs) which are biodegradable and biocompatible polymeric microspheres conveying cells on their biomimetic surface, therefore providing an adequate three-dimensional (3D) microenvironment. Moreover, they can release a growth factor in a prolonged manner. In order to implement ADSCs and PAMs for cardiac tissue engineering we first defined the biomimetic surface by studying the influence of matrix molecules laminin (LM) and fibronectin (FN), in combination with growth factors present in the cardiogenic niche, to further enhance the in vitro cardiac differentiation of ADSCs. We demonstrated that LM increased the expression of cardiac markers (Nkx2.5, GATA4, MEF2C) by ADSCs after 2 weeks in vitro. Interestingly, our results suggest that the 3D support provided by PAMs with a LM biomimetic surface (LM-PAMs) further enhanced the expression of cardiac markers and induced the expression of a more mature contractile protein, cardiac troponin I, compared with the 2D differentiating conditions after only 1 week in culture. The enrichment of the growth-factor cocktail with TGF-ß1 potentiated the cardiomyogenic differentiation. These results suggest that PAMs offering a LM biomimetic surface may be efficiently used for applications combining adult stem cells in tissue-engineering strategies of the ischemic heart.


Subject(s)
Adipose Tissue/cytology , Cell Lineage/drug effects , Coated Materials, Biocompatible/pharmacology , Laminin/pharmacology , Microspheres , Myocytes, Cardiac/cytology , Stem Cells/cytology , Biomarkers/metabolism , Cell Adhesion/drug effects , Cell Differentiation/drug effects , Cells, Cultured , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Fluorescent Antibody Technique , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Myocardium/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stem Cells/drug effects , Stem Cells/metabolism , Transforming Growth Factor beta1/pharmacology
4.
J Control Release ; 192: 82-94, 2014 Oct 28.
Article in English | MEDLINE | ID: mdl-24998940

ABSTRACT

The challenge of tissue engineering of the infarcted heart is how to improve stem cell engraftment, survival, homing, and differentiation for myocardial repair. We here propose to integrate human adipose-derived stem cells (ADSCs) and pharmacologically active microcarriers (PAMs), a three-dimensional (3D) carrier of cells and growth factors, into an injectable hydrogel (HG), to obtain a system that stimulates the survival and/or differentiation of the grafted cells toward a cardiac phenotype. PAMs are biodegradable and non-cytotoxic poly(lactic-co-glycolic acid) (PLGA) microspheres conveying cells on their 3D surface that deliver continuously and in a controlled manner a growth factor (GF) acting on the transported cells and on the microenvironment to improve engraftment. The choice of the appropriate GF and its protection during the formulation process and delivery are essential. In this study two GFs, hepatocyte growth factor (HGF) and insulin-like growth factor (IGF-1), have been encapsulated under a solid state in order to limit their interaction with the polymer and conserve their integrity. GF precipitation conditions and release profile from PAMs have been first investigated before combining them to ADSCs. The released IGF-1 and HGF induced the protein synthesis of cardiac differentiation markers GATA4, Nkx2.5, cTnI and CX43 after 1week in vitro. Moreover, the GFs accelerated cell cycle progression, as suggested by the increased expression of Cyclin D1 mRNA and the widespread distribution of Ki67 protein. Integrating PAMs within the thermosensitive P407 hydrogel increased their elastic properties but decreased the transcription of most cardiac markers. In contrast, CX43 expression increased in ADSC-PAM-GF complexes embedded within the hydrogel compared to the ADSCs cultured alone in the absence of P407. These results suggest that particulate scaffolds releasing HGF and IGF-1 may be beneficial for applications in tissue-engineering strategies for myocardial repair and the association with a P407 hydrogel can increase substrate elasticity and junction connections in ADSCs.


Subject(s)
Hepatocyte Growth Factor/administration & dosage , Hydrogel, Polyethylene Glycol Dimethacrylate/chemistry , Insulin-Like Growth Factor I/administration & dosage , Myocardium/cytology , Stem Cells/cytology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Adipose Tissue/cytology , Animals , Biomimetics , Cell Differentiation , Cells, Cultured , Human Umbilical Vein Endothelial Cells , Humans , Lactic Acid/chemistry , Mice , Models, Molecular , NIH 3T3 Cells , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Stem Cells/metabolism , Temperature
5.
J Cell Mol Med ; 17(1): 192-204, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23305078

ABSTRACT

Resistance of transplanted mesenchymal stem cells (MSCs) in post-ischemic heart is limited by their poor vitality. Vascular-endothelial-growth-factor-A (VEGF-A) as such or slowly released by fibronectin-coated pharmacologically-active-microcarriers (FN-PAM-VEGF) could differently affect survival kinases and anti-apoptotic mediator (e.g. Bcl-2). Therefore VEGF-A or FN-PAM-VEGF could differently enhance cell proliferation, and/or resistance to hypoxia/reoxygenation (H/R) of MSCs. To test these hypotheses MSCs were incubated for 6-days with VEGF-A alone or with FN-PAM-VEGF. In addition, MSCs pre-treated for 24-hrs with VEGF-A or FN-PAM-VEGF were subsequently exposed to H/R (72-hrs 3% O(2) and 3-hrs of reoxygenation). Cell-proliferation and post-hypoxic vitality were determined. Kinases were studied at 30-min., 1- and 3-days of treatment. Cell-proliferation increased about twofold (P < 0.01) 6-days after VEGF-A treatment, but by a lesser extent (55% increase) with FN-PAM-VEGF (P < 0.05). While MSC pre-treatment with VEGF-A confirmed cell-proliferation, pre-treatment with FN-PAM-VEGF protected MSCs against H/R. In the early phase of treatments, VEGF-A increased phospho-Akt, phospho-ERK-1/2 and phospho-PKCε compared to the untreated cells or FN-PAM-VEGF. Afterword, kinase phosphorylations were higher with VGEF, except for ERK-1/2, which was similarly increased by both treatments at 3 days. Only FN-PAM-VEGF significantly increased Bcl-2 levels. After H/R, lactate dehydrogenase release and cleaved Caspase-3 levels were mainly reduced by FN-PAM-VEGF. While VEGF-A enhances MSC proliferation in normoxia, FN-PAM-VEGF mainly hampers post-hypoxic MSC death. These different effects underscore the necessity of approaches suited to the various conditions. The use of FN-PAM-VEGF could be considered as a novel approach for enhancing MSC survival and regeneration in hostile environment of post-ischemic tissues.


Subject(s)
Lactic Acid/chemistry , Mesenchymal Stem Cells/drug effects , Polyglycolic Acid/chemistry , Vascular Endothelial Growth Factor A/pharmacology , Animals , Caspase 3/genetics , Caspase 3/metabolism , Cell Hypoxia , Cell Survival/drug effects , Drug Carriers , Drug Compounding , Kinetics , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Microspheres , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Oxygen/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer , Primary Cell Culture , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Vascular Endothelial Growth Factor A/administration & dosage , Vascular Endothelial Growth Factor A/chemistry
6.
Biomaterials ; 33(23): 5683-95, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22594970

ABSTRACT

An increasing number of studies in cardiac cell therapy have provided encouraging results for cardiac repair. Adult stem cells may overcome ethical and availability concerns, with the additional advantages, in some cases, to allow autologous grafts to be performed. However, the major problems of cell survival, cell fate determination and engraftment after transplantation, still remain. Tissue-engineering strategies combining scaffolds and cells have been developed and have to be adapted for each type of application to enhance stem cell function. Scaffold properties required for cardiac cell therapy are here discussed. New tissue engineering advances that may be implemented in combination with adult stem cells for myocardial infarction therapy are also presented. Biomaterials not only provide a 3D support for the cells but may also mimic the structural architecture of the heart. Using hydrogels or particulate systems, the biophysical and biochemical microenvironments of transplanted cells can also be controlled. Advances in biomaterial engineering have permitted the development of sophisticated drug-releasing materials with a biomimetic 3D support that allow a better control of the microenvironment of transplanted cells.


Subject(s)
Adult Stem Cells/cytology , Myocardial Infarction/therapy , Myocardium/cytology , Tissue Engineering/methods , Adult Stem Cells/transplantation , Animals , Drug Carriers/chemistry , Humans , Intercellular Signaling Peptides and Proteins/administration & dosage , Myocardial Infarction/surgery , Tissue Scaffolds/chemistry
7.
Eur J Pharm Biopharm ; 81(3): 609-16, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22561954

ABSTRACT

The regenerative potential of endothelial progenitor cell (EPC)-based therapies is limited due to poor cell viability and minimal retention following application. Neovascularization can be improved by means of scaffolds supporting EPCs. The aim of the present study was to investigate whether human early EPCs (eEPCs) could be efficiently cultured on pharmacologically active microcarriers (PAMs), made with poly(d,l-lactic-coglycolic acid) and coated with adhesion/extracellular matrix molecules. They may serve as a support for stem cells and may be used as cell carriers providing a controlled delivery of active protein such as the angiogenic factor, vascular endothelial growth factor-A (VEGF-A). eEPC adhesion to fibronectin-coated PAMs (FN-PAMs) was assessed by means of microscopic evaluation and by means of Alamar blue assay. Phospho ERK(1/2) and PARP-1 expression was measured by means of Western blot to assess the survival effects of FN-PAMs releasing VEGF-A (FN-VEGF-PAMs). The Alamar blue assay or a modified Boyden chamber assay was employed to assess proliferative or migratory capacity, respectively. Our data indicate that eEPCs were able to adhere to empty FN-PAMs within a few hours. FN-VEGF-PAMs increased the ability of eEPCs to adhere to them and strongly supported endothelial-like phenotype and cell survival. Moreover, the release of VEGF-A by FN-PAMs stimulated in vitro HUVEC migration and proliferation. These data strongly support the use of PAMs for supporting eEPC growth and survival and for stimulating resident mature human endothelial cells.


Subject(s)
Endothelial Cells/metabolism , Lactic Acid/chemistry , Polyglycolic Acid/chemistry , Stem Cells/metabolism , Tissue Scaffolds , Blotting, Western , Cell Adhesion , Cell Movement , Cell Proliferation , Cell Survival , Extracellular Matrix/metabolism , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neovascularization, Physiologic , Phosphorylation , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Polylactic Acid-Polyglycolic Acid Copolymer , Vascular Endothelial Growth Factor A/administration & dosage
8.
Eur J Pharm Sci ; 45(1-2): 128-37, 2012 Jan 23.
Article in English | MEDLINE | ID: mdl-22085679

ABSTRACT

A promising strategy to repair injured organs is possible by delivering a growth factor via poly-(d,l lactide-co-glycolide) (PLGA) microspheres; the latter are coated with adhesion molecules that serve as a support for cell delivery. At present, PLGA is not the optimal choice of polymer because of poor or incomplete protein release. The use of a more hydrophilic PLGA-PEG-PLGA (A-B-A) copolymer increases the degree of protein release. In this work, the impact of different combinations of (B) and (A) segments on the protein-release profile has been investigated. Continuous-release profiles, with no lag phases, were observed. The triblock ABA with a low molecular weight of PEG and a high molecular weight of PLGA showed an interesting release pattern with a small burst (<10% in 48 h) followed by sustained, protein release over 36 days. Incomplete protein release was found to be due to various causes: protein adsorption, protein aggregation and protein denaturation under acidic conditions. Interestingly, cell viability and cell adhesion on microspheres coated with fibronectin highlight the interest of these polymers for tissue engineering applications.


Subject(s)
Biomimetic Materials/chemistry , Bone Marrow Cells/cytology , Fibronectins/chemistry , Microspheres , Polyethylene Glycols/chemistry , Polyglactin 910/chemistry , Stromal Cells/cytology , Tissue Scaffolds/chemistry , Adsorption , Biomimetic Materials/metabolism , Bone Marrow Cells/metabolism , Cell Adhesion , Cell Survival , Cells, Cultured , Fibronectins/metabolism , Humans , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Molecular Weight , Poloxamer/chemistry , Polyethylene Glycols/metabolism , Polyglactin 910/metabolism , Protein Denaturation , Solubility , Stromal Cells/metabolism , Surface Properties , Tissue Engineering/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...