Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Int J Med Sci ; 16(1): 33-42, 2019.
Article in English | MEDLINE | ID: mdl-30662326

ABSTRACT

Background: α-Mangostin (αMG) is a natural substance that exerts a wide range of antitumor effects. Recently, we described that free αMG was able to dissociate multicellular tumour spheroids (MCTSs) generated from breast carcinoma cells and to reduce their cellular viability and motility. Here, αMG was encapsulated into lipidic nanoparticles (NPs), conjugated or not to a CD44 thioaptamer, and the anticancer action evaluated against MCF-7 breast MCTSs. Methods: NPs containing αMG were formulated with a core of polylactic-co-glycolyc acid. Some of them were decorated with a CD44 thioaptamer using as catalysts 1-ethyl-3- (3-dimethylaminopropyl) carbodiimide and N-hydroxysuccinimide. Both size and density of MCF-7-derived MCTSs were monitored during 72 h of treatment with NPs carrying 0.1, 0.5 and 1.0 µg/ml final concentrations of αMG. MCTSs were cultured on Matrigel or gelatine to better simulate the extracellular environment. Results: The NPs without thioaptamer and conveying 0.1 µg/ml αMG caused a significant dissociation of the MCTSs grown in gelatine after 24 h of treatment (p < 0.01). The most significant disaggregation of MCTSs was obtained using NPs carrying 0.5 µg/ml αMG (p < 0.01). A similar dissociating effect was observed when MCTSs were cultured in Matrigel under the same conditions for 48 - 72 h. By contrast, only concentrations over 1.0 µg/ml of free αMG were able to provoke a damage to MCTSs, consisting in a substantial reduction in their size (p < 0.05). Since the MCTS dissociation induced by αMG-loaded NPs occurred only in the presence of Matrigel or gelatine, an impairment of cell contacts to collagen fibres was likely responsible of this effect. Finally, the treatment of MCTSs with αMG-loaded NPs that were conjugated to the CD44 thioaptamer caused a similar decrease in density but a lower expansion of the spheroid, suggesting that a significant number of cells were died or arrested in cycle. Conclusion: Very low concentrations of αMG delivered by lipidic NPs are sufficient to provoke a substantial disaggregation of MCF-7 MCTSs that involves cell-to-collagen contacts. Similarly, the treatment of MCTSs with NPs conjugated to a CD44 thioaptamer leads to MCTS dissociation but through a more damaging action that causes also a reduction in cell number.


Subject(s)
Breast Neoplasms , Drug Delivery Systems , Hyaluronan Receptors , Nanoparticles , Protein Kinase Inhibitors/therapeutic use , Spheroids, Cellular/drug effects , Xanthones/therapeutic use , Aptamers, Nucleotide/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , MCF-7 Cells , Spheroids, Cellular/pathology
2.
Int J Med Sci ; 15(1): 23-30, 2018.
Article in English | MEDLINE | ID: mdl-29333084

ABSTRACT

Background: α-Mangostin (αMG) is extracted from Garcinia mangostana Linn and exerts antiproliferative activities. Although several researches on αMG were performed using cell monolayers, the in vitro pharmacological effects on 3D cancer models have never been investigated. Aim of the present study was to find new anticancer properties of αMG by evaluating the changes that this compound provokes in multicellular tumour spheroids (MCTSs). Methods: MCTSs were generated from MDA-MB-231 and MCF-7 breast tumour cell lines and then treated with 0.1÷30 µg/ml αMG for 24 and 48 h. MCTS size, density, and cell migration were determined by software elaboration of phase contrast images captured by a digital camera. Cell viability was evaluated by resazurin and acid phosphatase assays, while cell apoptosis was assessed by a fluorescent assay of caspase activity. The distribution of living cells inside MCTSs was shown by live/dead fluorescence staining. Results: A dose-dependent decrease in cell viability was obtained by treating MDA-MB-231 spheroids with αMG for 48 h (IC50 = 0.70-1.25 µg/ml). A significant reduction in spheroid volume, paralleled by its increased compactness, was observed only at concentration of 30 µg/ml, but not with lower doses of αMG. By contrast, αMG in the range of 5-15 µg/ml increased the size of MCTSs due to a parallel reduction in cell aggregation. The same window of concentrations was also able to stimulate cell apoptosis in a dose-dependent manner. Bimodal volumetric effects were also obtained by treating the spheroids generated from the MCF-7 cells with 0.1÷30 µg/ml αMG for 48 h. Finally, doses higher than 5 µg/ml caused a progressive impairment in cell migration from the edge of MDA-MB-231 MCTSs. Conclusion: After exposure at doses of αMG just above IC50, MDA-MB-231 spheroids showed a significant reduction in cell adhesion that did not stimulate cell migration but, on the contrary, blunted cell motility. These findings suggest a novel anticancer feature of αMG that could be taken into consideration to improve conventional drug penetration into the tumour bulk.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Garcinia mangostana/chemistry , Xanthones/pharmacology , Antineoplastic Agents/therapeutic use , Cell Aggregation/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Humans , Inhibitory Concentration 50 , Spheroids, Cellular/drug effects , Xanthones/therapeutic use
3.
Tissue Eng Part A ; 23(15-16): 811-822, 2017 08.
Article in English | MEDLINE | ID: mdl-28401805

ABSTRACT

At present, injuries or rupture of tendons are treated by surgical repair or conservative approaches with unpredictable clinical outcome. Alternative strategies to repair tendon defects without the undesirable side effects associated with the current options are needed. With this in mind, a tissue engineering approach has gained considerable attention as a promising strategy. Here we investigated a synthetic three-dimensional (3D) microenvironment able to interact with stem cells and inducing, via coupled biochemical and physical signals, their early commitment toward the tenogenic lineage. This multiphase 3D construct consisted of a braided hyaluronate elastic band merged with human bone marrow mesenchymal stem cells (hBMSCs) and poly-lactic-co-glycolic acid microcarriers loaded with human growth differentiation factor 5 (hGDF-5) by means of fibrin hydrogel. The multiphase structure allowed hBMSC culture under cyclic strain within a microenvironment where a controlled amount of hGDF-5 was regularly delivered. The cooperative biochemical and physical stimuli induced significantly increased expression of tenogenic markers, such as collagen type I and III, decorin, scleraxis, and tenascin-C, within only 3 days of dynamic hBMSC culture. This approach opens exciting perspectives for future development of engineered tendon tissue substitutes.


Subject(s)
Cell Lineage , Cellular Microenvironment , Growth Differentiation Factor 5/pharmacology , Mesenchymal Stem Cells/cytology , Stress, Mechanical , Tendons/cytology , Tissue Engineering/methods , Adult , Cell Lineage/drug effects , Elastic Modulus , Female , Gene Expression Regulation/drug effects , Humans , Male , Mesenchymal Stem Cells/drug effects , Microspheres , Tissue Scaffolds/chemistry
4.
Stem Cell Rev Rep ; 12(2): 189-201, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26661573

ABSTRACT

High tensile forces transmitted by tendons and ligaments make them susceptible to tearing or complete rupture. The present standard reparative technique is the surgical implantation of auto- or allografts, which often undergo failure.Currently, different cell types and biomaterials are used to design tissue engineered substitutes. Mechanical stimulation driven by dedicated devices can precondition these constructs to a remarkable degree, mimicking the local in vivo environment. A large number of dynamic culture instruments have been developed and many appealing results collected. Of the cells that have been used, tendon stem cells are the most promising for a reliable stretch-induced tenogenesis, but their reduced availability represents a serious limitation to upscaled production. Biomaterials used for scaffold fabrication include both biological molecules and synthetic polymers, the latter being improved by nanotechnologies which reproduce the architecture of native tendons. In addition to cell type and scaffold material, other variables which must be defined in mechanostimulation protocols are the amplitude, frequency, duration and direction of the applied strain. The ideal conditions seem to be those producing intermittent tension rather than continuous loading. In any case, all physical parameters must be adapted to the specific response of the cells used and the tensile properties of the scaffold. Tendon/ligament grafts in animals usually have the advantage of mechanical preconditioning, especially when uniaxial cyclic forces are applied to cells engineered into natural or decellularized scaffolds. However, due to the scarcity of in vivo research, standard protocols still need to be defined for clinical applications.


Subject(s)
Ligaments/cytology , Stem Cells/cytology , Tendons/cytology , Animals , Humans , Phenotype , Stress, Mechanical , Tissue Engineering/methods , Tissue Scaffolds
5.
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
6.
Med Hypotheses ; 84(3): 227-30, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25620576

ABSTRACT

Putative cancer stem cells (CSCs) reside in a hypoxic microenvironment where mesenchymal stem cells (MSCs) are also present. In this niche MSCs seem to promote the generation of CSCs and sustain tumor progression. Therefore, it may assume clinical relevance to produce a drug which kills not only CSCs but also MSCs. We hypothesized that bifunctional nanoparticles, loaded with a HIF-1α inhibitor and conjugated with an aptamer targeting a common receptor of CSCs and MSCs, may fulfill this strategy. The nanoparticle should ensure that: (1) the conveyed drug is less susceptible to degradation, (2) the common receptor of CSCs and MSCs is recognized by a superselective aptamer, and (3) receptor-mediated internalization is the main process to enter target cells. Small RNA or DNA aptamers represent an advantage over antibodies because do not cause immune reactions, are better internalized into the target cell, are more resistant to degradation, their cost of production are lower, and the purity of the oligonucleotide ligand is extremely elevated. Concerning the drugs to be delivered, we suggest to employ those exerting an anti-HIF-1α activity because they should be harmful for hypoxic CSCs and MCSs in their tumor niche but provide very limited toxicity, or even none, to well-oxygenated normal cells. Corresponding experimental approaches to perform pre-clinical studies and verify this hypothesis are also addressed.


Subject(s)
Cell Hypoxia/drug effects , Mesenchymal Stem Cells/metabolism , Models, Biological , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Neoplastic Stem Cells/metabolism , Tumor Microenvironment/drug effects , Aptamers, Peptide/metabolism , Aptamers, Peptide/therapeutic use , Drug Delivery Systems/methods , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Neoplasms/metabolism
7.
J. physiol. biochem ; 71(1): 141-153, mar. 2015.
Article in English | IBECS | ID: ibc-133911

ABSTRACT

Nitric oxide is an endogenous gas which exerts autocrine/paracrine actions by different signaling pathways and/or direct interactions with intracellular compounds and structures. Several processes are regulated by nitric oxide in stem cells including self-renewal, viability, migration, proliferation, and differentiation. The modulation of cell functions depends on its concentrations because opposite effects can be observed when low and high levels of nitric oxide are compared. Here, the responses to nitric oxide of adult stem/progenitor cells which are often used in regenerative medicine, including mesenchymal stem cells, hematopoietic stem cells, neural stem cells, endothelial progenitor cells, satellite cells, and fibro-adipogenic precursor cells, are reviewed. Therapeutic strategies which employ drugs releasing nitric oxide or modulating nitric oxide intracellular pathways are suggested to perform new ex vivo preconditioning or in vivo treatments suitable for stem/progenitor cell therapy and tissue engineering applications


Subject(s)
Humans , Nitric Oxide/pharmacokinetics , Adult Stem Cells/physiology , Paracrine Communication/physiology , Autocrine Communication/physiology , Mesenchymal Stem Cells/physiology , Hematopoietic Stem Cells/physiology , Ion Transport/physiology
8.
J Physiol Biochem ; 71(1): 141-53, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25526859

ABSTRACT

Nitric oxide is an endogenous gas which exerts autocrine/paracrine actions by different signaling pathways and/or direct interactions with intracellular compounds and structures. Several processes are regulated by nitric oxide in stem cells including self-renewal, viability, migration, proliferation, and differentiation. The modulation of cell functions depends on its concentrations because opposite effects can be observed when low and high levels of nitric oxide are compared. Here, the responses to nitric oxide of adult stem/progenitor cells which are often used in regenerative medicine, including mesenchymal stem cells, hematopoietic stem cells, neural stem cells, endothelial progenitor cells, satellite cells, and fibro-adipogenic precursor cells, are reviewed. Therapeutic strategies which employ drugs releasing nitric oxide or modulating nitric oxide intracellular pathways are suggested to perform new ex vivo preconditioning or in vivo treatments suitable for stem/progenitor cell therapy and tissue engineering applications.


Subject(s)
Adult Stem Cells/cytology , Cell Lineage , Mesenchymal Stem Cells/cytology , Nitric Oxide/physiology , Adult , Humans
9.
J Biomed Sci ; 21: 100, 2014 Oct 30.
Article in English | MEDLINE | ID: mdl-25358954

ABSTRACT

Hyaluronan (HA) is abundantly expressed in several human tissues and a variety of roles for HA has been highlighted. Particularly relevant for tissue repair, HA is actively produced during tissue injury, as widely evidenced in wound healing investigations. In the heart HA is involved in physiological functions, such as cardiac development during embryogenesis, and in pathological conditions including atherosclerosis and myocardial infarction. Moreover, owing to its relevant biological properties, HA has been widely used as a biomaterial for heart regeneration after a myocardial infarction. Indeed, HA and its derivatives are biodegradable and biocompatible, promote faster healing of injured tissues, and support cells in relevant processes including survival, proliferation, and differentiation. Injectable HA-based therapies for cardiovascular disease are gaining growing attention because of the benefits obtained in preclinical models of myocardial infarction. HA-based hydrogels, especially as a vehicle for stem cells, have been demonstrated to improve the process of cardiac repair by stimulating angiogenesis, reducing inflammation, and supporting local and grafted cells in their reparative functions. Solid-state HA-based scaffolds have been also investigated to produce constructs hosting mesenchymal stem cells or endothelial progenitor cells to be transplanted onto the infarcted surface of the heart. Finally, applying an ex-vivo mechanical stretching, stem cells grown in HA-based 3D scaffolds can further increase extracellular matrix production and proneness to differentiate into muscle phenotypes, thus suggesting a potential strategy to create a suitable engineered myocardial tissue for cardiac regeneration.


Subject(s)
Biocompatible Materials/therapeutic use , Heart/physiology , Hyaluronic Acid/therapeutic use , Myocardial Infarction/therapy , Regeneration , Angiogenesis Inducing Agents/metabolism , Angiogenesis Inducing Agents/pharmacology , Angiogenesis Inducing Agents/therapeutic use , Biocompatible Materials/metabolism , Biocompatible Materials/pharmacology , Humans , Hyaluronic Acid/metabolism , Hyaluronic Acid/pharmacology , Mesenchymal Stem Cells/metabolism , Wound Healing
10.
Stem Cells Int ; 2014: 434169, 2014.
Article in English | MEDLINE | ID: mdl-24511317

ABSTRACT

The production of a functional cardiac tissue to be transplanted in the injured area of the infarcted myocardium represents a challenge for regenerative medicine. Most cell-based grafts are unviable because of inadequate perfusion; therefore, prevascularization might be a suitable approach for myocardial tissue engineering. To this aim, cells with a differentiation potential towards vascular and cardiac muscle phenotypes have been cocultured in 2D or 3D appropriate scaffolds. In addition to these basic approaches, more sophisticated strategies have been followed employing mixed-cell sheets, microvascular modules, and inosculation from vascular explants. Technologies exerting spatial control of vascular cells, such as topographical surface roughening and ordered patterning, represent other ways to drive scaffold vascularization. Finally, microfluidic devices and bioreactors exerting mechanical stress have also been employed for high-throughput scaling-up production in order to accelerate muscle differentiation and speeding the endothelialization process. Future research should address issues such as how to optimize cells, biomaterials, and biochemical components to improve the vascular integration of the construct within the cardiac wall, satisfying the metabolic and functional needs of the myocardial tissue.

11.
Cardiovasc Res ; 102(1): 24-34, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24413773

ABSTRACT

AIMS: The ultimate cause of heart failure (HF) is not known to date. The cytoskeletal protein desmin is differentially modified and forms amyloid-like oligomers in HF. We postulated that desmin post-translational modifications (PTMs) could drive aberrant desmin aggregation in HF. Therefore, we identified these PTMs and investigated their impact on desmin amyloidogenicity in human and experimental HF. METHODS AND RESULTS: We detected increased levels of selectively phosphorylated and cleaved desmin in a canine pacing model of dyssynchronous HF (DHF) compared with either controls or animals treated with cardiac resynchronization therapy (CRT). This unique animal model combines clinically relevant features with the possibility of a partly rescued phenotype. We confirmed analogous changes in desmin modifications in human HF and identified two phosphorylation sites within a glycogen synthase kinase 3 (GSK3) consensus sequence. Desmin-positive oligomers were also increased in DHF hearts compared with controls. Their amyloid properties were decreased by treatment with CRT or an anti-amyloid small molecule. Finally, we confirmed GSK3's involvement with desmin phosphorylation using an in vitro model. CONCLUSIONS: Based on these findings, we postulate a new mechanism of cardiac toxicity based on the PTM-driven accumulation of desmin amyloid-like oligomers. Phosphorylation and cleavage as well as oligomers formation are reduced by treatment (CRT) indicating a relationship between the three. Finally, the decrease of desmin amyloid-like oligomers with CRT or small molecules points both to a general mechanism of HF based on desmin toxicity that is independent of protein mutations and to novel potential therapies.


Subject(s)
Desmin/metabolism , Heart Failure/metabolism , Protein Aggregates , Animals , Cardiac Resynchronization Therapy , Dogs , Glycogen Synthase Kinase 3/metabolism , Heart Failure/etiology , Mutation/genetics , Phosphorylation/physiology , Protein Processing, Post-Translational/physiology , Proteomics/methods
12.
J Tissue Eng Regen Med ; 8(10): 787-93, 2014 Oct.
Article in English | MEDLINE | ID: mdl-22865609

ABSTRACT

Much evidence in the literature demonstrates the effect of cyclic mechanical stretch in maintaining, or addressing, a muscle phenotype. Such results were obtained using several technical approaches, useful for the experimental collection of proofs of principle but probably unsuitable for application in clinical regenerative medicine. Here we aimed to design a reliable innovative bioreactor, acting as a stand-alone cell culture incubator, easy to operate and effective in addressing mesenchymal stem cells (MSCs) seeded onto a 3D bioreabsorbable scaffold, towards a muscle phenotype via the transfer of a controlled and highly-reproducible cyclic deformation. Electron microscopy, immunohistochemistry and biochemical analysis of the obtained pseudotissue constructs showed that cells 'trained' over 1 week: (a) displayed multilayer organization and invaded the 3D mesh of the scaffold; and (b) expressed typical markers of muscle cells. This effect was due only to physical stimulation of the cells, without the need of any other chemical or genetic manipulation. This device is thus proposed as a prototypal instrument to obtain pseudotissue constructs to test in cardiovascular regenerative medicine, using good manufacturing procedures.


Subject(s)
Bioreactors , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Mesenchymal Stem Cells/cytology , Tissue Scaffolds/chemistry , Animals , Cells, Cultured , Male , Mesenchymal Stem Cells/metabolism , Rats , Rats, Wistar
13.
Med Hypotheses ; 81(6): 1141-5, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24230458

ABSTRACT

In tumors intermittent hypoxia has been reported to be more representative than normoxia or continuous exposure to low oxygen concentrations. Intermittent hypoxia is thought to increase tumor resistance against both anti-cancer therapy and the sustained ischemia that randomly occurs because of the dynamic nature of tumor vasculature. Here, we hypothesize that the molecular mechanisms underlying intermittent hypoxia in tumor cells share some triggers, modulators, and end-effectors of the intermittent episodes of ischemia and reperfusion that characterize ischemic preconditioning and postconditioning. These are among the most effective maneuvers protecting cells from ischemia-reperfusion injury. If this hypothesis were confirmed, several well-investigated molecular mediators of pre/post-conditioning could be explored as therapeutic targets against tumor malignancy. For examples, drugs that completely block the cardioprotection induced by ischemic preconditioning, such as mitochondrial potassium ATP channel inhibitors or mitochondrial permeability transition pore openers, could be extraordinarily efficient in counteracting the adaptations of tumor cells and cancer stem cells to intermittent hypoxia. As a consequence, this strategy should be effective in blunting tumor capacity to progress toward malignancy and survive in ischemic conditions.


Subject(s)
Adaptation, Physiological/physiology , Cell Hypoxia/physiology , Ischemic Postconditioning/methods , Ischemic Preconditioning/methods , Models, Biological , Neoplasms/physiopathology , Humans , Neoplasms/drug therapy
14.
Biomed Res Int ; 2013: 918640, 2013.
Article in English | MEDLINE | ID: mdl-23936858

ABSTRACT

Owing to the inability of self-replacement by a damaged myocardium, alternative strategies to heart transplantation have been explored within the last decades and cardiac tissue engineering/regenerative medicine is among the present challenges in biomedical research. Hopefully, several studies witness the constant extension of the toolbox available to engineer a fully functional, contractile, and robust cardiac tissue using different combinations of cells, template bioscaffolds, and biophysical stimuli obtained by the use of specific bioreactors. Mechanical forces influence the growth and shape of every tissue in our body generating changes in intracellular biochemistry and gene expression. That is why bioreactors play a central role in the task of regenerating a complex tissue such as the myocardium. In the last fifteen years a large number of dynamic culture devices have been developed and many results have been collected. The aim of this brief review is to resume in a single streamlined paper the state of the art in this field.


Subject(s)
Heart/physiology , Myocardium/cytology , Regenerative Medicine , Tissue Engineering , Humans , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Stem Cells/cytology
15.
J Biomed Sci ; 20: 63, 2013 Aug 29.
Article in English | MEDLINE | ID: mdl-23985033

ABSTRACT

The efficiency of regenerative medicine can be ameliorated by improving the biological performances of stem cells before their transplantation. Several ex-vivo protocols of non-damaging cell hypoxia have been demonstrated to significantly increase survival, proliferation and post-engraftment differentiation potential of stem cells. The best results for priming cultured stem cells against a following, otherwise lethal, ischemic stress have been obtained with brief intermittent episodes of hypoxia, or anoxia, and reoxygenation in accordance with the extraordinary protection afforded by the conventional maneuver of ischemic preconditioning in severely ischemic organs. These protocols of hypoxic preconditioning can be rather easily reproduced in a laboratory; however, more suitable pharmacological interventions inducing stem cell responses similar to those activated in hypoxia are considered among the most promising solutions for future applications in cell therapy. Here we want to offer an up-to-date review of the molecular mechanisms translating hypoxia into beneficial events for regenerative medicine. To this aim the involvement of epigenetic modifications, microRNAs, and oxidative stress, mainly activated by hypoxia inducible factors, will be discussed. Stem cell adaptation to their natural hypoxic microenvironments (niche) in healthy and neoplastic tissues will be also considered.


Subject(s)
Adult Stem Cells/metabolism , Cell Differentiation , Regenerative Medicine/methods , Adult , Adult Stem Cells/cytology , Cell Hypoxia , Cell Survival , Humans , Ischemic Preconditioning, Myocardial
16.
Cell Biochem Biophys ; 67(2): 255-62, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23625166

ABSTRACT

Adipose-derived stem cells (ADSCs) are stromal mesenchymal stem cells isolated from lipoaspirates, and they display a broad potential to differentiate toward different lineages. The role of epigenetics in regulating the expression of their lineage-specific genes is under evaluation, however till date virtually nothing is known about the relative significance of cardiac-specific transcription factor genes in human ADSCs. The aim of this study was to investigate DNA promoter methylation and relevant histone modifications involving MEF-2C, GATA-4, and Nkx2.5 in native human ADSCs. CpG sites at the transcription start in their promoters were found unmethylated using methylation-specific PCR. Chromatin immunoprecipitation assay showed low levels of total acetylated H3 histone (acH3) and high levels of trimethylated lysine 27 in H3 histone (H3K27me3) which were associated with both GATA-4 and Nkx2.5 promoters, indicating their transcriptional repressive chromatin arrangement. On the other hand, the opposite was apparent for MEF-2C promoter. Accordingly, MEF-2C-but not GATA-4 and Nkx2.5-transcripts were evidenced in native human ADSCs. These results suggest that the chromatin arrangement of these early cardiac regulatory genes could be explored as a level of intervention to address the differentiation of human ADSCs toward the cardiac lineage.


Subject(s)
Adipose Tissue/cytology , Epigenesis, Genetic , Myocardium/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Transcription Factors/genetics , Adolescent , Adult , Chromatin/genetics , DNA Methylation , Female , Gene Expression Profiling , Histones/chemistry , Histones/genetics , Histones/metabolism , Humans , Lysine/metabolism , Male , Middle Aged , Phenotype , Promoter Regions, Genetic/genetics , Young Adult
17.
J Cell Mol Med ; 17(4): 518-30, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23480821

ABSTRACT

The aim of this study is to investigate the blood perfusion and the inflammatory response of the myocardial infarct area after transplanting a hyaluronan-based scaffold (HYAFF(®) 11) with bone marrow mesenchymal stem cells (MSCs). Nine-week-old female pigs were subjected to a permanent left anterior descending coronary artery ligation for 4 weeks. According to the kind of the graft, the swine subjected to myocardial infarction were divided into the HYAFF(®) 11, MSCs, HYAFF(®) 11/MSCs and untreated groups. The animals were killed 8 weeks after coronary ligation. Scar perfusion, evaluated by Contrast Enhanced Ultrasound echography, was doubled in the HYAFF(®) 11/MSCs group and was comparable with the perfusion of the healthy, non-infarcted hearts. The inflammation score of the MSCs and HYAFF(®) 11/MSCs groups was near null, revealing the role of the grafted MSCs in attenuating the cell infiltration, but not the foreign reaction strictly localized around the fibres of the scaffold. Apart from the inflammatory response, the native tissue positively interacted with the HYAFF(®) 11/MSCs construct modifying the extracellular matrix with a reduced presence of collagene and increased amount of proteoglycans. The border-zone cardiomyocytes also reacted favourably to the graft as a lower degree of cellular damage was found. This study demonstrates that the transplantation in the myocardial infarct area of autologous MSCs supported by a hyaluronan-based scaffold restores blood perfusion and almost completely abolishes the inflammatory process following an infarction. These beneficial effects are superior to those obtained after grafting only the scaffold or MSCs, suggesting that a synergic action was achieved using the cell-integrated polymer construct.


Subject(s)
Hyaluronic Acid/chemistry , Mesenchymal Stem Cell Transplantation , Myocardial Infarction/therapy , Neovascularization, Physiologic , Tissue Scaffolds , Animals , Cell Adhesion , Cell Shape , Cell Survival , Cells, Cultured , Coronary Vessels/physiopathology , Extracellular Matrix/metabolism , Female , Mesenchymal Stem Cells/physiology , Myocardial Infarction/physiopathology , Myocardium/pathology , Myocytes, Cardiac/physiology , Prostheses and Implants , Sus scrofa , Transplantation, Autologous
18.
J Surg Res ; 179(1): e21-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22475350

ABSTRACT

BACKGROUND: Permanence of grafted stem cells in the infarcted myocardial area has been suggested to be favored by tissue engineering strategies, including the application of a scaffold as a cell support. However, an estimation of how many cells remain localized in the site of transplantation has never been done. The aim of this work was to investigate the localization of mesenchymal stem cells (MSCs) grafted with a well cell-adhesive polymer in the scar region of the infarcted heart. MATERIALS AND METHODS: Rat MSCs were engineered in a hyaluronan-based scaffold (HYAFF(®)11) for 3 wk. The hearts of donor rats were also explanted, subjected to coronary artery ligation, and grafted into the abdomen of syngeneic rats. Two wk after coronary ligation a small dish of the HYAFF(®)11/MSC construct was introduced into a pouch created in the ventricular wall of the infarct area and left for 2 wk. RESULTS: Under ex vivo conditions, MSCs tightly adhered to the hyaluronan fibers and secreted abundant extracellular matrix. In contrast, HYAFF(®)11 was not more surrounded by the engrafted MSCs 2 wk after construct transplantation. Most MSCs migrated near the border zone of the infarcted area close to the coronary vessels. Moreover, the infarcted region of the heart was enriched in capillaries and the degree of fibrosis was attenuated. CONCLUSIONS: Two wk after transplantation most MSCs grafted in the infarcted myocardium with HYAFF(®)11 had left the scaffold and moved to the border zone. Nevertheless, this treatment increased the myocardial vascularization and reduced the degree of fibrosis in the scar area.


Subject(s)
Hyaluronic Acid , Mesenchymal Stem Cell Transplantation/methods , Myocardial Infarction/pathology , Myocardial Infarction/surgery , Tissue Engineering/methods , Tissue Scaffolds , Animals , Cell Adhesion/physiology , Cell Movement/physiology , Cells, Cultured , Cicatrix/pathology , Coronary Vessels/physiology , Disease Models, Animal , Endomyocardial Fibrosis/prevention & control , Male , Mesenchymal Stem Cells/pathology , Rats , Rats, Inbred Lew , Treatment Outcome
19.
Connect Tissue Res ; 54(1): 34-40, 2013.
Article in English | MEDLINE | ID: mdl-22853627

ABSTRACT

Adipose-derived stem cells (ASC) are usually isolated from lipoaspirates, but it is not known if the anesthetic solution injected into adipose tissue affects cell yield and functions. Two different samples were drawn from the abdominal region of female subjects. In the first, a physiological solution containing lidocaine/adrenaline was injected (wet liposuction, WL), while in the contralateral area, the sample was collected without injecting any solution (dry liposuction, DL). The aspirates were processed to investigate the yield of the stromal-vascular fraction (SVF) cells and ASC frequency, growth rate, apoptosis, and differentiation potential. The solid dried mass of fresh WL isolates was lower than that of DL isolates (p < 0.01) due to the presence, in the former, of a liquid solution. As a consequence, the amount of WL-SVF cells was 18.7% lower than those obtained from DL (p < 0.01); this difference was also observed under culture conditions. In addition, the number of colony-forming unit-fibroblasts (CFU-Fs) obtained from 1 × 10(3) SVF cells was 25.5% lower in WL-aspirates than DL-aspirates (p < 0.05) owing, at least in part, to the observed presence of ASC [corrected] in the liquid solution of the WL isolates. After WL and DL, no differences were observed in ASC growth rate, apoptosis, or differentiation potential toward adipogenic, osteogenic, and endothelial cell lineages. In conclusion, WL yields about 40% fewer ASC than DL due to the combined effect of tissue dilution and the reduced frequency of ASC in the SVF. The main biological features of ASC are suitable for cell-based therapies.


Subject(s)
Adipose Tissue/cytology , Adult Stem Cells/cytology , Lipectomy/methods , Multipotent Stem Cells/cytology , Tissue and Organ Harvesting/methods , Adipocytes/cytology , Adipocytes/metabolism , Adolescent , Adult , Apoptosis , Cell Count , Cell Differentiation , Cell Proliferation , Cell Survival , Cells, Cultured , Colony-Forming Units Assay , Female , Humans , Middle Aged , Young Adult
20.
Oncol Rep ; 28(6): 2271-7, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22992787

ABSTRACT

We previously described a cohort of grade II oligodendroglioma (OII) patients, in whom the loss of heterozygosity (LOH) 19q was present in the subgroup at a higher risk of relapse. In this study, we evaluated the CpG methylation of the putative tumor suppressor epithelial membrane protein 3 (EMP3, 19q13.3) gene promoter in the same OII cohort, to investigate whether a correlation could be found between EMP3 cytogenetic and epigenetic loss and higher risk of relapse. Twenty-three tumor samples from OII patients were collected over a period of 10 years. Seventeen glioblastoma (GBM) samples (2 of which were relapses) were collected from 15 patients. The EMP3, O6-methylguanine methyltransferase (MGMT) and cyclooxygenase 2 (COX2) promoter methylation, evaluated by methylation-specific PCR, and the isocitrate dehydrogenase 1 (IDH1) mutation, identified by sequencing, were compared between the OII and GBM histotypes. The EMP3 promoter methylation was correlated with the analysis of LOH 19q, performed by microsatellite amplification, in OII patients. Disease progression-free interval was evaluated in the OII patients with the EMP3 methylation with either LOH 19q or conserved chromosome 19 arms. The EMP3 and MGMT promoter methylation was more frequent in OII than in GBM patients, and the IDH1 mutation was absent in GBM. The COX2 promoter was unmethylated in both histotypes. Both LOH+/- 19q OII patients showed EMP3 hypermethylation. Concomitant LOH 19q and EMP3 gene promoter methylation was observed in the OII patients at a higher risk of relapse. Our results suggest that a total (cytogenetic and epigenetic) functional loss of both EMP3 alleles accounts for the reduced disease progression-free interval in OII patients. Although the small sample size limits the strength of this study, our results support testing this hypothesis in larger cohorts of patients, considering the methylation of the EMP3 gene promoter together with LOH 19q as an indication for treatment with adjuvant therapy ab initio in order to improve the overall survival of OII patients.


Subject(s)
Brain Neoplasms/genetics , Chromosomes, Human, Pair 19/genetics , DNA Methylation , Loss of Heterozygosity , Membrane Glycoproteins/genetics , Oligodendroglioma/genetics , Adult , Aged , Brain Neoplasms/metabolism , CpG Islands , Cyclooxygenase 2/genetics , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Male , Membrane Glycoproteins/metabolism , Middle Aged , O(6)-Methylguanine-DNA Methyltransferase/genetics , Oligodendroglioma/metabolism , Promoter Regions, Genetic/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...