Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
1.
Br J Pharmacol ; 177(20): 4683-4700, 2020 10.
Article in English | MEDLINE | ID: mdl-32757222

ABSTRACT

BACKGROUND AND PURPOSE: Prenatal caffeine exposure (PCE) can cause developmental toxicity of long bones in offspring, but the long-term effects and the underlying mechanism have not been fully clarified. Here, we investigated the effects of PCE peak bone mass accumulation and osteoporosis susceptibility in offspring and its intrauterine programming mechanism. EXPERIMENTAL APPROACH: Pregnant Wistar rats were administrated intragastrically with saline or caffeine (120 mg·kg-1 ·day-1 ) on gestational days 9-20. The serum and bone samples were collected from the fetal and postnatal offspring for bone mass, genes expression and corticosterone analysis. Then, rat bone marrow mesenchymal stem cells (BMSCs) were treated with corticosterone in vitro to confirm the molecular mechanism. KEY RESULTS: PCE caused fetal bone dysplasia in male and female offspring. In adulthood, PCE reduced peak bone mass and increased osteoporosis susceptibility in male offspring but not in females. Meanwhile, PCE only decreased the H3K9ac and expression levels of 11ß-hydroxysteroid dehydrogenase 2 (11ß-HSD2) before and after birth in the male offspring but not in the females. Moreover, the high level of corticosterone induced by PCE down-regulated the H3K9ac and expression levels of 11ß-HSD2 through promoting glucocorticoid receptor (GR; NR3C1) into the nucleus of bone marrow mesenchymal stem cells (BMSCs) and recruiting histone deacetylase 11 (HDAC11) binding to 11ß-HSD2 promoter region, which further enhanced the effect of corticosterone on suppressing osteogenic function of BMSCs. CONCLUSION AND IMPLICATIONS: PCE caused osteoporosis susceptibility in male adult offspring, which attributed to the low-functional programming of 11ß-HSD2 induced by corticosterone via GR/HDAC11 signalling.


Subject(s)
Osteoporosis , Prenatal Exposure Delayed Effects , 11-beta-Hydroxysteroid Dehydrogenase Type 2 , 11-beta-Hydroxysteroid Dehydrogenases , Animals , Caffeine/toxicity , Female , Glucocorticoids , Male , Osteoporosis/chemically induced , Pregnancy , Rats , Rats, Wistar
2.
J Tissue Eng Regen Med ; 12(2): 360-369, 2018 02.
Article in English | MEDLINE | ID: mdl-28486755

ABSTRACT

Scaffolds laden with stem cells are a promising approach for articular cartilage repair. Investigations have shown that implantation of artificial matrices, growth factors or chondrocytes can stimulate cartilage formation, but no existing strategies apply mechanical stimulation on stratified scaffolds to mimic the cartilage environment. The purpose of this study was to adapt a spraying method for stratified cartilage engineering and to stimulate the biosubstitute. Human mesenchymal stem cells from bone marrow were seeded in an alginate (Alg)/hyaluronic acid (HA) or Alg/hydroxyapatite (Hap) gel to direct cartilage and hypertrophic cartilage/subchondral bone differentiation, respectively, in different layers within a single scaffold. Homogeneous or composite stratified scaffolds were cultured for 28 days and cell viability and differentiation were assessed. The heterogeneous scaffold was stimulated daily. The mechanical behaviour of the stratified scaffolds were investigated by plane-strain compression tests. Results showed that the spraying process did not affect cell viability. Moreover, cell differentiation driven by the microenvironment was increased with loading: in the layer with Alg/HA, a specific extracellular matrix of cartilage, composed of glycosaminoglycans and type II collagen was observed, and in the Alg/Hap layer more collagen X was detected. Hap seemed to drive cells to a hypertrophic chondrocytic phenotype and increased mechanical resistance of the scaffold. In conclusion, mechanical stimulations will allow for the production of a stratified biosubstitute, laden with human mesenchymal stem cells from bone marrow, which is capable in vivo to mimic all depths of chondral defects, thanks to an efficient combination of stem cells, biomaterial compositions and mechanical loading.


Subject(s)
Cell Differentiation , Mesenchymal Stem Cells/cytology , Stress, Mechanical , Tissue Scaffolds/chemistry , Aged , Alginates/pharmacology , Biocompatible Materials/pharmacology , Cell Differentiation/drug effects , Chondrogenesis/drug effects , Durapatite/pharmacology , Humans , Hydrogel, Polyethylene Glycol Dimethacrylate/pharmacology , Mesenchymal Stem Cells/drug effects , Middle Aged
3.
Immunol Res ; 65(6): 1130-1138, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29019081

ABSTRACT

The ability of natural killer (NK) cells to kill tumor cells without antigen recognition makes them appealing as an adoptive immunotherapy. However, NK cells are not routinely used in the context of leukemic relapse after hematopoietic stem cell transplantation. Patients who experience relapse can be treated with donor lymphocyte infusions (DLI) based on small-cell fractions frozen at the time of transplantation. Since peripheral blood stem cells (PBSCs) are increasingly used as a stem cell source and as a source of cells for DLI, we aimed to evaluate the impact of G-SCF mobilization on NK cell phenotype, subset repartition, and functionality. Immunomagnetically isolated NK cells from healthy donor blood, donor PBSCs, and patient PBSCs were expanded for 14 days with IL-15. The expansion capacity, phenotype, and functions (cytokine secretion and cytotoxicity) of NK cell subsets based on CD56 and CD16 expression were then evaluated. Mobilized sources showed a significant decrease of CD56brightCD16+ NK cells (28 versus 74%), whereas a significant increase (64 versus 15%) of CD56brightCD16- NK cells was observed in comparison with peripheral blood. Patient-mobilized NK cells showed a significantly decreased cytotoxicity, and antibody-dependent cell cytototoxicity (ADCC) was also observed to a lesser extent in NK cells from healthy donor PBSC. G-CSF-mobilized NK cell TNF-α and IFN-γ secretion was impaired at day 0 compared to healthy donors but was progressively restored after culture. In conclusion, expansion of NK cells from G-CSF-mobilized sources may progressively improve their functionality.


Subject(s)
Granulocyte Colony-Stimulating Factor/metabolism , Hematopoietic Stem Cells/physiology , Immunotherapy, Adoptive/methods , Interleukin-15/metabolism , Killer Cells, Natural/physiology , Lymphocyte Subsets/physiology , Neoplasms/immunology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Cytotoxicity, Immunologic , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cell Transplantation , Humans , Immunologic Surveillance , Immunomagnetic Separation , Killer Cells, Natural/transplantation , Leukapheresis , Lymphocyte Activation , Lymphocyte Subsets/transplantation , Transplantation, Homologous
4.
Stem Cell Res Ther ; 8(1): 161, 2017 07 05.
Article in English | MEDLINE | ID: mdl-28676126

ABSTRACT

BACKGROUND: The umbilical cord is becoming a notable alternative to bone marrow (BM) as a source of mesenchymal stromal cells (MSC). Although age-dependent variations in BM-MSC are well described, less data are available for MSC isolated from Wharton's jelly (WJ-MSC). We initiated a study to identify whether obstetric factors influenced MSC properties. We aimed to evaluate the correlation between a large number of obstetric factors collected during pregnancy and until peripartum (related to the mother, the labor and delivery, and the newborn) with WJ-MSC proliferation and chondrogenic differentiation parameters. METHODS: Correlations were made between 27 obstetric factors and 8 biological indicators including doubling time at passage (P)1 and P2, the percentage of proteoglycans and collagens, and the relative transcriptional expression of Sox-9, aggrecans, and total type 2 collagen (Coll2T). RESULTS: Amongst the obstetric factors considered, birth weight, the number of amenorrhea weeks, placental weight, normal pregnancy, and the absence of preeclampsia were identified as relevant factors for cell expansion, using multivariate linear regression analysis. Since all the above parameters are related to term, we concluded that WJ-MSC from healthy, full-term infants exhibit greater proliferation capacity. As for chondrogenesis, we also observed that obstetric factors influencing proliferation seemed beneficial, with no negative impact on MSC differentiation. CONCLUSIONS: Awareness of obstetric factors influencing the proliferation and/or differentiation of WJ-MSC will make it possible to define criteria for collecting optimal umbilical cords with the aim of decreasing the variability of WJ-MSC batches produced for clinical use in cell and tissue engineering.


Subject(s)
Amenorrhea , Birth Weight , Cell Differentiation , Cell Proliferation , Chondrogenesis , Mesenchymal Stem Cells/metabolism , Umbilical Cord/metabolism , Adult , Collagen Type II/metabolism , Female , Humans , Mesenchymal Stem Cells/cytology , Pregnancy , Risk Factors , SOX9 Transcription Factor/metabolism , Umbilical Cord/cytology
5.
Biomed Mater Eng ; 28(s1): S87-S94, 2017.
Article in English | MEDLINE | ID: mdl-28372282

ABSTRACT

The construction of the high biocompatible biomaterials pretreated with MSC offers a promising strategy to improve the effects of stem cell therapy for the myocardial infarction (MI). However, assembling vascularized three-dimensional (3-D) myocardial tissues remains an enormous challenge. In this study, we optimized the decellularization protocol with the umbilical artery to construct microporous 3-D scaffold which is suitable for the stem cells (SC) proliferation. The SD rats underwent proximal left coronary ligation and a 5-mm diameter microporous SC patch was implanted directly on the infarct area (SC patch group). The LV contractile function, regional myocardial wall compliance, and tissue histology were assessed 4 weeks after patch implantation. The MSC patch integrated to the local heart tissue and the neo-vessels have been observed in the MSC patch. The vessels in the MSC patch were positive for the CD31 (marker for the mature endothelial cells). The left ventricle wall was thicker in the MSC patch group than the control group (p<0.05 vs. empty patch group). And the LVEF has been improved in the MSC patch group than empty patch group (59±6.7% vs. 31±4.5%, p<0.05). CONCLUSIONS: Our results showed that the implantation of the MSC patch improved cardiac contractile function in heart infarction rat model. The construction of artificial tissue from the decellularized umbilical artery and the MSC may open a promising perspective for the tissue therapy for MI.


Subject(s)
Heart/physiopathology , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Myocardial Infarction/therapy , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Umbilical Arteries/transplantation , Animals , Heart Ventricles/cytology , Heart Ventricles/physiopathology , Male , Myocardial Infarction/physiopathology , Rats, Sprague-Dawley , Umbilical Arteries/chemistry , Umbilical Arteries/ultrastructure
6.
Biomed Mater Eng ; 28(s1): S113-S119, 2017.
Article in English | MEDLINE | ID: mdl-28372286

ABSTRACT

Liver transplantation is the definitive treatment for patients with end-stage liver diseases (ESLD). However, it is hampered by shortage of liver donor. Liver tissue engineering, aiming at fabricating new livers in vitro, provides a potential resolution for donor shortage. Three elements need to be considered in liver tissue engineering: seeding cell resources, scaffolds and bioreactors. Studies have shown potential cell sources as hepatocytes, hepatic cell line, mesenchymal stem cells and others. They need scaffolds with perfect biocompatiblity, suitable micro-structure and appropriate degradation rate, which are essential charateristics for cell attachment, proliferation and secretion in forming extracellular matrix. The most promising scaffolds in research include decellularized whole liver, collagens and biocompatible plastic. The development and function of cells in scaffold need a microenvironment which can provide them with oxygen, nutrition, growth factors, et al. Bioreactor is expected to fulfill these requirements by mimicking the living condition in vivo. Although there is great progress in these three domains, a large gap stays still between their researches and applications. Herein, we summarized the recent development in these three major fields which are indispensable in liver tissue engineering.


Subject(s)
Biocompatible Materials/chemistry , Hepatocytes/cytology , Liver/cytology , Mesenchymal Stem Cells/cytology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Bioreactors , Humans , Liver/growth & development , Liver, Artificial
7.
Biomed Mater Eng ; 28(s1): S229-S235, 2017.
Article in English | MEDLINE | ID: mdl-28372299

ABSTRACT

In tissue engineering approaches, the quality of substitutes is a key element to determine its ability to treat cartilage defects. However, in clinical practice, the evaluation of tissue-engineered cartilage substitute quality is not possible due to the invasiveness of the standard procedure, which is to date histology. The aim of this work was to validate a new innovative system performed from two-photon excitation laser adapted to an optical macroscope to evaluate at macroscopic scale the collagen network in cartilage tissue-engineered substitutes in confrontation with gold standard histologic techniques or immunohistochemistry to visualize type II collagen. This system permitted to differentiate the quality of collagen network between ITS and TGF-ß1 treatments. Multiscale large field imaging combined to multimodality approaches (SHG-TCSPC) at macroscopical scale represent an innovative and non-invasive technique to monitor the quality of collagen network in cartilage tissue-engineered substitutes before in vivo implantation.


Subject(s)
Cartilage/anatomy & histology , Chondrocytes/cytology , Collagen Type II/analysis , Mesenchymal Stem Cells/cytology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Cartilage/chemistry , Cartilage/cytology , Cartilage/growth & development , Chondrocytes/metabolism , Chondrogenesis , Humans , Mesenchymal Stem Cells/metabolism , Transforming Growth Factor beta1/administration & dosage , Transforming Growth Factor beta1/metabolism
8.
Immunol Cell Biol ; 94(4): 342-56, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26510892

ABSTRACT

Wharton's jelly mesenchymal stromal cells (WJ-MSCs) are promising candidates for tissue engineering, as their immunomodulatory activity allows them to escape immune recognition and to suppress several immune cell functions. To date, however, few studies have investigated the effect of differentiation of the MSCs on this immunomodulation. To address this question, we sought to determine the impact of differentiation toward endothelial cells on immunoregulation by WJ-MSCs. Following differentiation, the endothelial-like cells (ELCs) were positive for CD31, vascular endothelial cadherin and vascular endothelial growth factor receptor 2, and able to take up acetylated low-density lipoproteins. The expression of HLA-DR and CD86, which contribute to MSCs immunoprivilege, was still weak after differentiation. We then co-cultured un- and differentiated MSCs with immune cells, under conditions of both direct and indirect contact. The proliferation and phenotype of the immune cells were analyzed and the mediators secreted by both ELCs and WJ-MSCs quantified. Interleukin (IL)-6, IL-1ß, prostaglandin E2 and in particular indoleamine-2,3-dioxygenase expression were upregulated in ELCs on stimulation by T and NK cells, suggesting the possible involvement of these factors in allosuppression. ELCs co-cultured with T cells were able to generate CD25(+) T cells, which were shown to be of the CD4(+)CD25(+)FoxP3(+) regulatory subset. Direct contact between NK cells and ELCs or WJ-MSCs decreased the level of NK-activating receptor natural-killer group 2, member D. Moreover, direct co-culturing with ELCs stimulates CD73 acquisition on NK cells, a mechanism which may induce adenosine secretion by the cells and lead to an immunosuppressive function. Taken together, our results show that ELCs obtained following differentiation of WJ-MSCs remain largely immunosuppressive.


Subject(s)
Endothelial Cells/physiology , Killer Cells, Natural/physiology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , T-Lymphocytes, Regulatory/physiology , 5'-Nucleotidase/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Coculture Techniques , Cytokines/metabolism , Female , Forkhead Transcription Factors/metabolism , Humans , Immunosuppression Therapy , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism
9.
J Immunother ; 39(1): 27-35, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26641259

ABSTRACT

Adoptive antiviral cellular immunotherapy by infusion of virus-specific T cells (VSTs) is becoming an alternative treatment for viral infection after hematopoietic stem cell transplantation. The T memory stem cell (TSCM) subset was recently described as exhibiting self-renewal and multipotency properties which are required for sustained efficacy in vivo. We wondered if such a crucial subset for immunotherapy was present in VSTs. We identified, by flow cytometry, TSCM in adenovirus (ADV)-specific interferon (IFN)-γ+ T cells before and after IFN-γ-based immunomagnetic selection, and analyzed the distribution of the main T-cell subsets in VSTs: naive T cells (TN), TSCM, T central memory cells (TCM), T effector memory cell (TEM), and effector T cells (TEFF). In this study all of the different T-cell subsets were observed in the blood sample from healthy donor ADV-VSTs, both before and after IFN-γ-based immunomagnetic selection. As the IFN-γ-based immunomagnetic selection system sorts mainly the most differentiated T-cell subsets, we observed that TEM was always the major T-cell subset of ADV-specific T cells after immunomagnetic isolation and especially after expansion in vitro. Comparing T-cell subpopulation profiles before and after in vitro expansion, we observed that in vitro cell culture with interleukin-2 resulted in a significant expansion of TN-like, TCM, TEM, and TEFF subsets in CD4IFN-γ T cells and of TCM and TEM subsets only in CD8IFN-γ T cells. We demonstrated the presence of all T-cell subsets in IFN-γ VSTs including the TSCM subpopulation, although this was weakly selected by the IFN-γ-based immunomagnetic selection system.


Subject(s)
Adenoviridae/immunology , Interferon-gamma/metabolism , Lymphocyte Count , T-Cell Antigen Receptor Specificity/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Adenoviridae Infections/immunology , Adenoviridae Infections/therapy , Antigens, Surface/metabolism , Cell Culture Techniques , Cytotoxicity, Immunologic , Healthy Volunteers , Humans , Immunologic Memory , Immunomagnetic Separation , Immunophenotyping , Immunotherapy, Adoptive , Phenotype
10.
Stem Cell Res Ther ; 6: 260, 2015 Dec 30.
Article in English | MEDLINE | ID: mdl-26718750

ABSTRACT

BACKGROUND: Due to their intrinsic properties, stem cells are promising tools for new developments in tissue engineering and particularly for cartilage tissue regeneration. Although mesenchymal stromal/stem cells from bone marrow (BM-MSC) have long been the most used stem cell source in cartilage tissue engineering, they have certain limits. Thanks to their properties such as low immunogenicity and particularly chondrogenic differentiation potential, mesenchymal stromal/stem cells from Wharton's jelly (WJ-MSC) promise to be an interesting source of MSC for cartilage tissue engineering. METHODS: In this study, we propose to evaluate chondrogenic potential of WJ-MSC embedded in alginate/hyaluronic acid hydrogel over 28 days. Hydrogels were constructed by the original spraying method. Our main objective was to evaluate chondrogenic differentiation of WJ-MSC on three-dimensional scaffolds, without adding growth factors, at transcript and protein levels. We compared the results to those obtained from standard BM-MSC. RESULTS: After 3 days of culture, WJ-MSC seemed to be adapted to their new three-dimensional environment without any detectable damage. From day 14 and up to 28 days, the proportion of WJ-MSC CD73(+), CD90(+), CD105(+) and CD166(+) decreased significantly compared to monolayer marker expression. Moreover, WJ-MSC and BM-MSC showed different phenotype profiles. After 28 days of scaffold culture, our results showed strong upregulation of cartilage-specific transcript expression. WJ-MSC exhibited greater type II collagen synthesis than BM-MSC at both transcript and protein levels. Furthermore, our work highlighted a relevant result showing that WJ-MSC expressed Runx2 and type X collagen at lower levels than BM-MSC. CONCLUSIONS: Once seeded in the hydrogel scaffold, WJ-MSC and BM-MSC have different profiles of chondrogenic differentiation at both the phenotypic level and matrix synthesis. After 4 weeks, WJ-MSC, embedded in a three-dimensional environment, were able to adapt to their environment and express specific cartilage-related genes and matrix proteins. Today, WJ-MSC represent a real alternative source of stem cells for cartilage tissue engineering.


Subject(s)
Hydrogels/chemistry , Mesenchymal Stem Cells/cytology , Tissue Engineering , Adult , Alginates/chemistry , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cartilage/physiology , Cell Differentiation , Cell Survival , Cells, Cultured , Chondrogenesis , Collagen Type II/metabolism , Collagen Type X/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Humans , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Middle Aged , Phenotype , Regeneration , Wharton Jelly/cytology , Wharton Jelly/metabolism
12.
Biomed Mater Eng ; 25(1 Suppl): 27-39, 2015.
Article in English | MEDLINE | ID: mdl-25538053

ABSTRACT

INTRODUCTION: Stem cells for autologous and allogenic transplantation are obtained from several sources including bone marrow, peripheral blood or cord blood. Accurate enumeration of viable CD34+ hematopoietic stem cells (HSC) is routinely used in clinical settings, especially to monitor progenitor cell mobilization and apheresis. The number of viable CD34+ HSC has also been shown to be the most critical factor in haematopoietic engraftment. The International Society for Cellular Therapy actually recommends the use of single-platform flow cytometry system using 7-AAD as a viability dye. AIM: In a way to move routine analysis from a BD FACSCaliburTM instrument to a BD FACSCantoTM II, according to ISO 15189 standard guidelines, we define laboratory performance data of the BDTM Stem Cell Enumeration (SCE) kit on a CE-IVD system including a BD FACSCanto II flow cytometer and the BD FACSCantoTM Clinical Software. InterQCTM software, a real time internet laboratory QC management system developed by VitroTM and distributed by Becton DickinsonTM, was also tested to monitor daily QC data, to define the internal laboratory statistics and to compare them to external laboratories. METHODS: Precision was evaluated with BDTM Stem Cell Control (high and low) results and the InterQC software, an internet laboratory QC management system by Vitro. This last one drew Levey-Jennings curves and generated numeral statistical parameters allowing detection of potential changes in the system performances as well as interlaboratory comparisons. Repeatability, linearity and lower limits of detection were obtained with routine samples from different origins. Agreement evaluation between BD FACSCanto II system versus BD FACSCalibur system was tested on fresh peripheral blood, freeze-thawed apheresis, fresh bone marrow and fresh cord blood samples. RESULTS: Instrument's measure and staining repeatability clearly evidenced acceptable variability on the different samples tested. Intra- and inter-laboratory CV in CD34+ cell absolute count are consistent and reproducible. Linearity analysis, established between 2 and 329 cells/µl showed a linear relation between expected counts and measured counts (R2=0.97). Linear regression and Bland-Altman representations showed an excellent correlation on samples from different sources between the two systems and allowed the transfer of routine analysis from BD FACSCalibur to BD FACSCanto II. CONCLUSIONS: The BD SCE kit provides an accurate measure of the CD34 HSC, and can be used in daily routine to optimize the enumeration of hematopoietic CD34+ stem cells by flow cytometry. Moreover, the InterQC system seems to be a very useful tool for laboratory daily quality monitoring and thus for accreditation.


Subject(s)
Cell Count/standards , Flow Cytometry/standards , Guidelines as Topic , Hematopoietic Stem Cell Transplantation/standards , Hematopoietic Stem Cells/cytology , Antigens, CD34/immunology , Cell Count/methods , Cells, Cultured , France , Hematopoietic Stem Cells/immunology , Humans , Internationality , Reproducibility of Results , Sensitivity and Specificity , Stem Cell Transplantation/standards
13.
Biomed Mater Eng ; 25(1 Suppl): 41-6, 2015.
Article in English | MEDLINE | ID: mdl-25538054

ABSTRACT

Under physiological conditions, there is a production of limited range of free radicals. However, when the cellular antioxidant defence systems, overwhelm and fail to reverse back the free radicals to their normal basal levels, there is a creation of a condition of redox disequilibrium termed "oxidative stress", which is implicated in a very wide spectrum of genetic, metabolic, and cellular responses. The excess of free radicals can, cause unfavourable molecular alterations to biomolecules through oxidation of lipids, proteins, RNA and DNA, that can in turn lead to mutagenesis, carcinogenesis, and aging. Mesenchymal stem cells (MSCs) have been proven to be a promising source of cells for regenerative medicine, and to be useful in the treatment of pathologies in which tissue damage is linked to oxidative stress. Moreover, MSCs appeared to efficiently manage oxidative stress and to be more resistant to oxidative insult than normal somatic cells, making them an interesting and testable model for the role of oxidative stress in the aging process. In addition, aging is accompanied by a progressive decline in stem cell function, resulting in less effective tissue homeostasis and repair. Also, there is an obvious link between intracellular reactive oxygen species levels and cellular senescence. To date, few studies have investigated the promotion of aging by oxidative stress on human MSCs, and the mechanism by which oxidative stress induce stem cell aging is poorly understood. In this context, the aim of this review is to gain insight the current knowledge about the molecular mechanisms of aging-induced oxidative stress in human MSCs.


Subject(s)
Aging/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Cell Proliferation/physiology , Cell Survival/physiology , Cells, Cultured , Humans
14.
Biomed Mater Eng ; 25(1 Suppl): 65-71, 2015.
Article in English | MEDLINE | ID: mdl-25538057

ABSTRACT

The scaffolds prepared from the tissue decellularization conserve the porous 3-D structure and provide an optimal matrix for the tissue regeneration. Since decade, the enzymatic digestion, chemical reagent treatment and mechanical actions such as eversion and abrasion have been used to remove the cells from the intact matrix. In this study, we optimized an enzymatic method to decellularize the umbilical artery to construct a 3-D porous scaffold which is suitable for the culture of mesenchymal stem cells (MSCs). The scaffold maintained the interconnected porous structure. It remained the similar high water content 95.3 ± 1% compared to 94.9 ± 0.6% in the intact umbilical artery (p>0.05). The decellularization process decreased the stress from 0.24 ± 0.05 mPa to 0.15 ± 0.06 mPa (p<0.05). However the decellularization did not change the strain of the artery (45 ± 15% vs. 53 ± 10%, p>0.05). When the scaffold was transplanted to the subcutaneous tissue in the wild type mice, there were less T cells appeared in the surrounding tissue which meant the decreased the immunogenicity by decellularization. This scaffold also supported the adhesion and proliferation of the MSCs. In this study, we constructed a biological compatible porous scaffold from the decellularized umbilical artery which may provide a suitable scaffold for cell-matrix interaction studies and for tissue engineering.


Subject(s)
Biocompatible Materials/chemical synthesis , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Tissue Scaffolds , Umbilical Arteries/chemistry , Animals , Cell Adhesion/physiology , Cell Proliferation/physiology , Cell-Free System , Cells, Cultured , Compressive Strength , Elastic Modulus , Equipment Design , Equipment Failure Analysis , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Humans , Materials Testing , Mice , Porosity , Tensile Strength , Tissue Engineering/instrumentation , Umbilical Arteries/metabolism
15.
Biomed Mater Eng ; 25(1 Suppl): 79-85, 2015.
Article in English | MEDLINE | ID: mdl-25538059

ABSTRACT

Designing unique nanostructured biomimetic materials is a new challenge in modern regenerative medicine. In order to develop functional substitutes for damaged organs or tissues, several methods have been used to create implants able to regenerate robust and durable bone. Electrospinning produces nonwoven scaffolds based on polymer nanofibers mimicking the fibrillar organization of bone extracellular matrix. Here, we describe a biomimetic 3D thick nanofibrous scaffold obtained by electrospinning of the biodegradable, bioresorbable and FDA-approved polymer, poly(ε-caprolactone). Such scaffold presents a thickness reaching one centimeter. We report here the demonstration that the designed nanostructured implant is able to induce in vivo bone regeneration.


Subject(s)
Bone Regeneration/physiology , Bone Substitutes/chemical synthesis , Nanofibers/chemistry , Osteoblasts/physiology , Skull Fractures/therapy , Tissue Scaffolds , Animals , Biomimetic Materials/chemical synthesis , Cells, Cultured , Equipment Design , Equipment Failure Analysis , Extracellular Matrix/chemistry , Humans , Materials Testing , Mice , Nanofibers/ultrastructure , Osteoblasts/cytology , Osteoblasts/transplantation , Osteogenesis/physiology , Particle Size , Polyesters/chemistry , Skull Fractures/pathology , Skull Fractures/physiopathology , Treatment Outcome
16.
Biomed Mater Eng ; 25(1 Suppl): 111-9, 2015.
Article in English | MEDLINE | ID: mdl-25538062

ABSTRACT

X-linked Hypohidrotic Ectodermal Dysplasia (XLHED) is associated to a large spectrum of ectodermal and extra-ectodermal symptoms, especially craniofacial bone morphological, structural and metabolic anomalies. This skeletal phenotype described in affected patients and in the Ta mutant mouse model leads to craniofacial dysmorphies, endosseous implants and jaw bone grafts complications. Bone tissue bioengineering based on the use of PCL synthetic nanofibrous membrane and BMP nanoreservoirs appears as an original and promising approach to prevent such complications in the context of dysfunctional bone. Use of osteoblasts or stem cells seeded biomembranes appears as another strategy developed on the Tabby (Ta) model of XLHED. The Ta mouse experimental model is used to study the jaw bone response during the post-operative period after bone lesion and placement of synthetic PCL membrane functionalized with nanoreservoirs embedding different BMPs dimers or seeded with living cells.


Subject(s)
Bone Diseases, Developmental/therapy , Bone Regeneration/physiology , Disease Models, Animal , Ectodermal Dysplasia 1, Anhidrotic/therapy , Stem Cell Transplantation/methods , Animals , Bone Diseases, Developmental/genetics , Bone Diseases, Developmental/pathology , Ectodermal Dysplasia 1, Anhidrotic/pathology , Ectodysplasins/genetics , Forecasting , Mice , Mutation/genetics , Regenerative Medicine/methods , Regenerative Medicine/trends , Stem Cell Transplantation/trends
17.
Biomed Mater Eng ; 25(1 Suppl): 137-43, 2015.
Article in English | MEDLINE | ID: mdl-25538064

ABSTRACT

The shortage of organ resource has been limiting the application of liver transplantation. Bioartificial liver construction is increasingly focused as a replacement treatment. To product a bioartificial liver, three elements must be considered: seeding cells, scaffold and bioreactor. Recent studies have shown that several methods can successfully differentiate MSC (mesenchymal stem cells) derived from Wharton's jelly into hepatocyte, such as stimulating MSC by cytokines and growth factors, direct and indirect co-culture MSC with hepatocytes, or promote MSC differentiation by 3-dimensional matrix. In some cases, differentiation of MSC into hepatocytes can also be an alternative approach for whole organ transplantation in treatment of acute and chronic liver diseases. In this review, the characterization of MSC from Wharton's jelly, their potential of application in liver tissue engineering on base of decellularized scaffold, their status of banking and their preclinical work performed will be discussed.


Subject(s)
Liver, Artificial , Mesenchymal Stem Cells/cytology , Organ Culture Techniques/instrumentation , Tissue Engineering/instrumentation , Tissue Scaffolds , Wharton Jelly/cytology , Bioreactors , Cell Differentiation/physiology , Cells, Cultured , Humans , Mesenchymal Stem Cell Transplantation/instrumentation , Mesenchymal Stem Cells/physiology , Prosthesis Design
18.
Biomed Mater Eng ; 25(1 Suppl): 159-66, 2015.
Article in English | MEDLINE | ID: mdl-25538066

ABSTRACT

OBJECTIVES: In present study, we plan to produce a decellularization protocol from rat liver to generate a three-dimensional whole organ scaffold. METHODS: A combination of 1% SDS and 1% tritonX-100 were used orderly to decellularize rat livers. After about 6 h of interactive antegrade/retrograde perfusion, a decellularized whole translucent liver scaffold with integrated blood vessel networks was generated. The decellularized livers are charactered by light microscopy, scanning electron microscopy, and biochemical analysis (DNA quantification) for preservation of the three-dimension of extracellular matrix architecture. RESULTS: The decellularization protocol was verified by observation of the whole translucent liver organ with intact vascular trees under macroscopy, in conjunction with the hematoxylin-eosin staining that showed no cells or nuclear material remained. Additionally, the Masson's stain indicted that the extracellular proteins were well kept and scanning electron microscopy (SEM) revealed a preserved decellularized matrix architecture. Compared to normal livers, DNA in the decellularized livers was quantified less than 10% at the same mass. CONCLUSIONS: The current method of decellularization protocol was feasible, simple and quick, and was verified by an absence of residual cells. The decellularized extracellular matrix had preserved integrate vascular network and a three-dimensional architecture.


Subject(s)
Extracellular Matrix/chemistry , Liver, Artificial , Liver/chemistry , Liver/ultrastructure , Tissue Engineering/instrumentation , Tissue Scaffolds , Animals , Cell-Free System , Equipment Design , Equipment Failure Analysis , Extracellular Matrix/ultrastructure , Female , Rats , Rats, Sprague-Dawley
19.
Biomed Mater Eng ; 25(1 Suppl): 187-97, 2015.
Article in English | MEDLINE | ID: mdl-25538069

ABSTRACT

BACKGROUND: Non-union of long bones is still a current problem in traumatology. Although corticocancellous bone autograft remains the usual procedure for the treatment of non-union, innovative therapies such as, percutaneous autologous concentrated bone marrow grafting (PABMG), are now appearing. MATERIAL AND METHODS: Over a period of 8 years, 45 non-union of long bones were treated by PABMG in the Department of Orthopaedic and Traumatologic Surgery (University Hospital of Nancy, France): 26 tibiae, 16 femurs, 3 humeri. Efficiency was evaluated by clinical criteria: full weight-bearing without pain, absence of motion at non-union site, and radiological criteria: healing of 3 corticales out of 4. RESULTS: Eighteen out of 28 non-unions at the tibia were healed (69%), 10 at the femur (63%), but none was noticed at the humerus. Some pejorative prognosis factors were noted such as: tobacco, alcohol abuse, diabetes and history of infection at the fracture site. An earlier grafting improved the success rate. The number of CFU-F (Colony Forming Unit Fibroblastic) affected the healing time more than the healing rate. CONCLUSION: The procedure, even though a little invasive, enables the healing of non-union in two out of three cases with less morbidity than conventional procedures. This procedure fits perfectly into the therapeutic arsenal of non-union.


Subject(s)
Bone Marrow Transplantation/methods , Fractures, Malunited/diagnostic imaging , Fractures, Malunited/therapy , Adolescent , Adult , Aged , Aged, 80 and over , Female , Humans , Injections , Male , Middle Aged , Radiography , Treatment Outcome , Young Adult
20.
Biomed Mater Eng ; 24(1 Suppl): 47-52, 2014.
Article in English | MEDLINE | ID: mdl-24928917

ABSTRACT

While mesenchymal stem cells represent an interesting cell source for regenerative medicine, several points have to be investigated to improve their use in clinical, and in particular in the elderly population. This work studied the proliferation capacity of mesenchymal stem cells isolated from human bone marrow in function of donor's age. Doubling time after in vitro culture, clonogenicity and phenotype were analyzed in 17 samples ranging from 3 to 85 years old (mean 47 ± 27). Results showed an increase in the doubling time for cell coming from old donor compared to cells coming from young ones. This was accompanied by a decrease in clonogenicity while no changes were observe in cell phenotype. In conclusion, this study showed an effect of donor's age on the proliferation capacity of mesenchymal stem cells isolated from bone marrow that was correlated to a decrease in clonogenicity. The comprehension of molecular mechanism involved in this process could help to improve the clinical application of mesenchymal stem cells.


Subject(s)
Age Factors , Cell Proliferation/physiology , Colony-Forming Units Assay/methods , Mesenchymal Stem Cells , Adolescent , Adult , Aged , Aged, 80 and over , Bone Marrow Cells/cytology , Cells, Cultured , Child , Child, Preschool , Humans , Middle Aged , Regenerative Medicine , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...