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.
J Appl Physiol (1985) ; 132(2): 375-387, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34941443

ABSTRACT

Increases in positive end-expiratory pressure (PEEP) or recruitment maneuvers may increase stress in lung parenchyma, extracellular matrix, and lung vessels; however, adaptative responses may occur. We evaluated the effects of PEEP on lung damage and cardiac function when increased abruptly, gradually, or more gradually in experimental mild/moderate acute respiratory distress syndrome (ARDS) induced by Escherichia coli lipopolysaccharide intratracheally. After 24 h, Wistar rats (n = 48) were randomly assigned to four mechanical ventilation strategies according to PEEP levels: 1) 3 cmH2O for 2 h (control); 2) 3 cmH2O for 1 h followed by an abrupt increase to 9 cmH2O for 1 h (no adaptation time); 3) 3 cmH2O for 30 min followed by a gradual increase to 9 cmH2O over 30 min then kept constant for 1 h (shorter adaptation time); and 4) more gradual increase in PEEP from 3 cmH2O to 9 cmH2O over 1 h and kept constant thereafter (longer adaptation time). At the end of the experiment, oxygenation improved in the shorter and longer adaptation time groups compared with the no-adaptation and control groups. Diffuse alveolar damage and expressions of interleukin-6, club cell protein-16, vascular cell adhesion molecule-1, amphiregulin, decorin, and syndecan were higher in no adaptation time compared with other groups. Pulmonary arterial pressure was lower in longer adaptation time than in no adaptation (P = 0.002) and shorter adaptation time (P = 0.025) groups. In this model, gradually increasing PEEP limited lung damage and release of biomarkers associated with lung epithelial/endothelial cell and extracellular matrix damage, as well as the PEEP-associated increase in pulmonary arterial pressure.NEW & NOTEWORTHY In a rat model of Escherichia coli lipopolysaccharide-induced mild/moderate acute respiratory distress syndrome, a gradual PEEP increase (shorter adaptation time) effectively mitigated histological lung injury and biomarker release associated with lung inflammation, damage to epithelial cells, endothelial cells, and the extracellular matrix compared with an abrupt increase in PEEP. A more gradual PEEP increase (longer adaptation time) decreased lung damage, pulmonary vessel compression, and pulmonary arterial pressure.


Subject(s)
Endothelial Cells , Respiratory Distress Syndrome , Animals , Rats , Lung , Positive-Pressure Respiration , Rats, Wistar , Respiratory Distress Syndrome/therapy
2.
Tissue Eng Part A ; 27(3-4): 256-269, 2021 02.
Article in English | MEDLINE | ID: mdl-32580647

ABSTRACT

Stem cell-replacement therapies have been proposed as a potential tool to treat sensorineural hearing loss by aiding the regeneration of spiral ganglion neurons (SGNs) in the inner ear. However, transplantation procedures have yet to be explored thoroughly to ensure proper cell differentiation and optimal transplant procedures. We hypothesized that the aggregation of human embryonic stem cell (hESC)-derived otic neuronal progenitor (ONP) cells into a multicellular form would improve their function and their survival in vivo post-transplantation. We generated hESC-derived ONP spheroids-an aggregate form conducive to differentiation, transplantation, and prolonged cell survival-to optimize conditions for their transplantation. Our findings indicate that these cell spheroids maintain the molecular and functional characteristics similar to those of ONP cells, which are upstream in the SGN lineage. Moreover, our phenotypical, electrophysiological, and mechanical data suggest an optimal spheroid transplantation point after 7 days of in vitro three-dimensional (3D) culture. We have also developed a feasible transplantation protocol for these spheroids using a micropipette aided by a digital microinjection system. In summary, the present work demonstrates that the transplantation of ONP cells in spheroid form into the inner ear through micropipette 7 days after seeding for 3D spheroid culture is an expedient and viable method for stem cell replacement therapies in the inner ear.


Subject(s)
Human Embryonic Stem Cells , Cell Differentiation , Humans , Neurons , Spheroids, Cellular , Spiral Ganglion , Stem Cell Transplantation
3.
Stem Cells Transl Med ; 9(10): 1244-1256, 2020 10.
Article in English | MEDLINE | ID: mdl-32538526

ABSTRACT

Silicosis is a pneumoconiosis caused by inhaled crystalline silica microparticles, which trigger inflammatory responses and granuloma formation in pulmonary parenchyma, thus affecting lung function. Although systemic administration of mesenchymal stromal cells (MSCs) ameliorates lung inflammation and attenuates fibrosis in experimental silicosis, it does not reverse collagen deposition and granuloma formation. In an attempt to improve the beneficial effects of MSCs, magnetic targeting (MT) has arisen as a potential means of prolonging MSC retention in the lungs. In this study, MSCs were incubated with magnetic nanoparticles and magnets were used for in vitro guidance of these magnetized MSCs and to enhance their retention in the lungs in vivo. In vitro assays indicated that MT improved MSC transmigration and expression of chemokine receptors. In vivo, animals implanted with magnets for 48 hours had significantly more magnetized MSCs in the lungs, suggesting improved MSC retention. Seven days after magnet removal, silicotic animals treated with magnetized MSCs and magnets showed significant reductions in static lung elastance, resistive pressure, and granuloma area. In conclusion, MT is a viable technique to prolong MSC retention in the lungs, enhancing their beneficial effects on experimentally induced silicosis. MT may be a promising strategy for enhancing MSC therapies for chronic lung diseases.


Subject(s)
Lung/pathology , Magnetics/methods , Mesenchymal Stem Cells/pathology , Nanoparticles/metabolism , Silicosis/therapy , Animals , Disease Models, Animal , Female , Humans , Mice , Silicosis/physiopathology
4.
Front Immunol ; 9: 1147, 2018.
Article in English | MEDLINE | ID: mdl-29881388

ABSTRACT

Asthma is characterized by chronic lung inflammation and airway hyperresponsiveness. Despite recent advances in the understanding of its pathophysiology, asthma remains a major public health problem and, at present, there are no effective interventions capable of reversing airway remodeling. Mesenchymal stromal cell (MSC)-based therapy mitigates lung inflammation in experimental allergic asthma; however, its ability to reduce airway remodeling is limited. We aimed to investigate whether pre-treatment with eicosapentaenoic acid (EPA) potentiates the therapeutic properties of MSCs in experimental allergic asthma. Seventy-two C57BL/6 mice were used. House dust mite (HDM) extract was intranasally administered to induce severe allergic asthma in mice. Unstimulated or EPA-stimulated MSCs were administered intratracheally 24 h after final HDM challenge. Lung mechanics, histology, protein levels of biomarkers, and cellularity in bronchoalveolar lavage fluid (BALF), thymus, lymph nodes, and bone marrow were analyzed. Furthermore, the effects of EPA on lipid body formation and secretion of resolvin-D1 (RvD1), prostaglandin E2 (PGE2), interleukin (IL)-10, and transforming growth factor (TGF)-ß1 by MSCs were evaluated in vitro. EPA-stimulated MSCs, compared to unstimulated MSCs, yielded greater therapeutic effects by further reducing bronchoconstriction, alveolar collapse, total cell counts (in BALF, bone marrow, and lymph nodes), and collagen fiber content in airways, while increasing IL-10 levels in BALF and M2 macrophage counts in lungs. In conclusion, EPA potentiated MSC-based therapy in experimental allergic asthma, leading to increased secretion of pro-resolution and anti-inflammatory mediators (RvD1, PGE2, IL-10, and TGF-ß), modulation of macrophages toward an anti-inflammatory phenotype, and reduction in the remodeling process. Taken together, these modifications may explain the greater improvement in lung mechanics obtained. This may be a promising novel strategy to potentiate MSCs effects.


Subject(s)
Asthma/metabolism , Eicosapentaenoic Acid/pharmacology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Animals , Asthma/etiology , Asthma/pathology , Asthma/therapy , Biomarkers , Bone Marrow/immunology , Bone Marrow/metabolism , Bone Marrow/pathology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cytokines/metabolism , Female , Immunohistochemistry , Inflammation Mediators/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Macrophage Activation/immunology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Male , Mesenchymal Stem Cell Transplantation/methods , Mice , Mucus/metabolism , Thymus Gland/immunology , Thymus Gland/metabolism
5.
Nanomedicine ; 14(7): 2075-2085, 2018 10.
Article in English | MEDLINE | ID: mdl-29933023

ABSTRACT

The magnetic targeting (MT) technique improves delivery of mesenchymal stromal cells (MSCs) to target sites. However, the moderate-intensity static magnetic fields (SMF) used for MT may exert adverse effects on MSCs. Thus, we aimed to evaluate the effects of SMF on MSCs in vitro. Cells were initially magnetized using citrate-coated magnetite nanoparticles. Then, control and magnetized MSCs were transferred to an in vitro MT system and exposed to 0.3-0.45 Tesla SMFs. MSC viability, morphology, ultrastructure, proliferation rates, differentiation, and immunomodulation were evaluated after 24 and 48 hours of exposure. MSCs temporarily lost viability and exhibited ultrastructural changes after exposure to SMFs, regardless of magnetization. Moreover, exposure to SMF reduced magnetized MSC proliferation rates. Nevertheless, MSCs remained functional (i.e., capable of differentiating, secreting repair mediators, and modulating alveolar macrophage phenotype). Thus, the experimental protocol tested in this experiment can be applied in future in vivo MT studies.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Macrophages, Alveolar/immunology , Magnetic Fields , Magnetite Nanoparticles/administration & dosage , Mesenchymal Stem Cells/cytology , Animals , Cells, Cultured , Macrophages, Alveolar/drug effects , Magnetite Nanoparticles/chemistry , Mesenchymal Stem Cells/drug effects , Mice , Mice, Inbred C57BL
6.
Stem Cell Res Ther ; 9(1): 45, 2018 02 26.
Article in English | MEDLINE | ID: mdl-29482654

ABSTRACT

Due to their anti-inflammatory, antiapoptotic, antimicrobial, and antifibrotic properties, mesenchymal stromal cells (MSCs) have been considered a promising alternative for treatment of respiratory diseases. Nevertheless, even though MSC administration has been demonstrated to be safe in clinical trials, to date, few studies have shown evidence of MSC efficacy in respiratory diseases. The present review describes strategies to enhance the beneficial effects of MSCs, including preconditioning (under hypoxia, oxidative stress, heat shock, serum deprivation, and exposure to inflammatory biological samples) and genetic manipulation. These strategies can variably promote increases in MSC survival rates, by inducing expression of cytoprotective genes, as well as increase MSC potency by improving secretion of reparative factors. Furthermore, these strategies have been demonstrated to enhance the beneficial effects of MSCs in preclinical lung disease models. However, there is still a long way to go before such strategies can be translated from bench to bedside.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Respiratory Tract Diseases , Animals , Clinical Trials as Topic , Disease Models, Animal , Humans , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Respiratory Tract Diseases/metabolism , Respiratory Tract Diseases/pathology , Respiratory Tract Diseases/therapy
7.
Stem Cell Res Ther ; 8(1): 58, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28279201

ABSTRACT

Mesenchymal stromal cells (MSCs) have been extensively investigated in the field of regenerative medicine. It is known that the success of MSC-based therapies depends primarily on effective cell delivery to the target site where they will secrete vesicles and soluble factors with immunomodulatory and potentially reparative properties. However, some lesions are located in sites that are difficult to access, such as the heart, spinal cord, and joints. Additionally, low MSC retention at target sites makes cell therapy short-lasting and, therefore, less effective. In this context, the magnetic targeting technique has emerged as a new strategy to aid delivery, increase retention, and enhance the effects of MSCs. This approach uses magnetic nanoparticles to magnetize MSCs and static magnetic fields to guide them in vivo, thus promoting more focused, effective, and lasting retention of MSCs at the target site. In the present review, we discuss the magnetic targeting technique, its principles, and the materials most commonly used; we also discuss its potential for MSC enhancement, and safety concerns that should be addressed before it can be applied in clinical practice.


Subject(s)
Immunomodulation , Magnetite Nanoparticles/therapeutic use , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Humans , Magnetic Fields , Magnetite Nanoparticles/chemistry , Regenerative Medicine
8.
J Nanobiotechnology ; 14(1): 59, 2016 Jul 18.
Article in English | MEDLINE | ID: mdl-27431051

ABSTRACT

BACKGROUND: Nanoparticles' unique features have been highly explored in cellular therapies. However, nanoparticles can be cytotoxic. The cytotoxicity can be overcome by coating the nanoparticles with an appropriated surface modification. Nanoparticle coating influences biocompatibility between nanoparticles and cells and may affect some cell properties. Here, we evaluated the biocompatibility of gold and maghemite nanoparticles functionalized with 2,3-dimercaptosuccinic acid (DMSA), Au-DMSA and γ-Fe2O3-DMSA respectively, with human mesenchymal stem cells. Also, we tested these nanoparticles as tracers for mesenchymal stem cells in vivo tracking by computed tomography and as agents for mesenchymal stem cells magnetic targeting. RESULTS: Significant cell death was not observed in MTT, Trypan Blue and light microscopy analyses. However, ultra-structural alterations as swollen and degenerated mitochondria, high amounts of myelin figures and structures similar to apoptotic bodies were detected in some mesenchymal stem cells. Au-DMSA and γ-Fe2O3-DMSA labeling did not affect mesenchymal stem cells adipogenesis and osteogenesis differentiation, proliferation rates or lymphocyte suppression capability. The uptake measurements indicated that both inorganic nanoparticles were well uptaken by mesenchymal stem cells. However, Au-DMSA could not be detected in microtomograph after being incorporated by mesenchymal stem cells. γ-Fe2O3-DMSA labeled cells were magnetically responsive in vitro and after infused in vivo in an experimental model of lung silicosis. CONCLUSION: In terms of biocompatibility, the use of γ-Fe2O3-DMSA and Au-DMSA as tracers for mesenchymal stem cells was assured. However, Au-DMSA shown to be not suitable for visualization and tracking of these cells in vivo by standard computed microtomography. Otherwise, γ-Fe2O3-DMSA shows to be a promising agent for mesenchymal stem cells magnetic targeting.


Subject(s)
Cell Tracking/methods , Magnetite Nanoparticles/chemistry , Mesenchymal Stem Cells/drug effects , Silicosis/diagnostic imaging , Staining and Labeling/methods , Succimer/pharmacology , Adolescent , Adult , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dental Pulp/cytology , Dental Pulp/drug effects , Ferric Compounds/chemistry , Ferric Compounds/pharmacology , Gold/chemistry , Gold/pharmacology , Humans , Magnetite Nanoparticles/ultrastructure , Mesenchymal Stem Cells/ultrastructure , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/ultrastructure , Particle Size , Primary Cell Culture , Silicosis/pathology , Succimer/chemistry , X-Ray Microtomography
SELECTION OF CITATIONS
SEARCH DETAIL
...