Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
J Cardiovasc Dev Dis ; 10(2)2023 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-36826582

RESUMO

Therapy microencapsulation allows minimally invasive, safe, and effective administration. Hepatocyte growth factor (HGF) has angiogenic, anti-inflammatory, anti-apoptotic, and anti-fibrotic properties. Our objective was to evaluate the cardiac safety and effectiveness of intracoronary (IC) administration of HGF-loaded extended release microspheres in an acute myocardial infarction (AMI) swine model. An IC infusion of 5 × 106 HGF-loaded microspheres (MS+HGF, n = 7), 5 × 106 placebo microspheres (MS, n = 7), or saline (SAL, n = 7) was performed two days after AMI. TIMI flow and Troponin I (TnI) values were assessed pre- and post-treatment. Cardiac function was evaluated with magnetic resonance imaging (cMR) before injection and at 10 weeks. Plasma cytokines were determined to evaluate the inflammatory profile and hearts were subjected to histopathological evaluation. Post-treatment coronary flow was impaired in five animals (MS+HGF and MS group) without significant increases in TnI. One animal (MS group) died during treatment. There were no significant differences between groups in cMR parameters at any time (p > 0.05). No statistically significant changes were found between groups neither in cytokines nor in histological analyses. The IC administration of 5 × 106 HGF-loaded-microspheres 48 h post-AMI did not improve cardiac function, nor did it decrease inflammation or cardiac fibrosis in this experimental setting.

2.
Front Pharmacol ; 11: 01332, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33101010

RESUMO

More than a century has passed since the first surgical mesh for hernia repair was developed, and, to date, this is still the most widely used method despite the great number of complications it poses. The purpose of this study was to combine stem cell therapy and laparoscopy for the treatment of congenital hernia in a swine animal model. Porcine bone marrow-derived mesenchymal stem cells (MSCs) were seeded on polypropylene surgical meshes using a fibrin sealant solution as a vehicle. Meshes with (cell group) or without (control group) MSCs were implanted through laparoscopy in Large White pigs with congenital abdominal hernia after the approximation of hernia borders (implantation day). A successive laparoscopic biopsy of the mesh and its surrounding tissues was performed a week after implantation, and surgical meshes were excised a month after implantation. Ultrasonography was used to measure hernia sizes. Flow cytometry, histological, and gene expression analyses of the biopsy and necropsy samples were performed. The fibrin sealant solution was easy to prepare and preserved the viability of MSCs in the surgical meshes. Ultrasonography demonstrated a significant reduction in hernia size 1 week after implantation in the cell group relative to that on the day of implantation (p < 0.05). Flow cytometry of the mesh-infiltrated cells showed a non-significant increase of M2 macrophages when the cell group was compared with the control group 1 week after implantation. A significant decrease in the gene expression of VEGF and a significant increase in TNF expression were determined in the cell group 1 month after implantation compared with gene expressions in the control group (p < 0.05). Here, we propose an easy and feasible method to combine stem cell therapy and minimally invasive surgical techniques for hernia repair. In this study, stem cell therapy did not show a great immunomodulatory or regenerative effect in overcoming hernia-related complications. However, our clinically relevant animal model with congenital hernia closely resembles the clinical human condition. Further studies should be focused on this valuable animal model to evaluate stem cell therapies in hernia surgery.

3.
Stem Cell Rev Rep ; 16(3): 626, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32107730

RESUMO

The original version of this article unfortunately contained a mistake. In the author group, the correct family name of Dr. Rebeca is "Blázquez" and the correct family name of Dr. Francisco Miguel is "Sánchez-Margallo."

4.
Stem Cell Rev Rep ; 16(3): 612-625, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31865532

RESUMO

Acute myocardial infarction triggers a strong inflammatory response in the affected cardiac tissue. New therapeutic tools based on stem cell therapy may modulate the unbalanced inflammation in the damaged cardiac tissue, contributing to the resolution of this pathological condition. The main goal of this study was to analyze the immunomodulatory effects of cardiosphere-derived cells (CDCs) and their extracellular vesicles (EV-CDCs), delivered by intrapericardial administration in a clinically relevant animal model, during the initial pro-inflammatory phase of an induced myocardial infarction. This effect was assessed in peripheral blood and pericardial fluid leukocytes from infarcted animals. Additionally, cardiac functional parameters, troponin I, hematological and biochemical components were also analyzed to characterize myocardial infarction-induced changes, as well as the safety aspects of these procedures. Our preclinical study demonstrated a successful myocardial infarction induction in all animals, without any reported adverse effect related to the intrapericardial administration of CDCs or EV-CDCs. Significant changes were observed in biochemical and immunological parameters after myocardial infarction. The analysis of peripheral blood leukocytes revealed an increase of M2 monocytes in the EV-CDCs group, while no differences were reported in other lymphocyte subsets. Moreover, arginase-1 (M2-differentiation marker) was significantly increased in pericardial fluids 24 h after EV-CDCs administration. In summary, we demonstrate that, in our experimental conditions, intrapericardially administered EV-CDCs have an immunomodulatory effect on monocyte polarization, showing a beneficial effect for counteracting an unbalanced inflammatory reaction in the acute phase of myocardial infarction. These M2 monocytes have been defined as "pro-regenerative cells" with a pro-angiogenic and anti-inflammatory activity.


Assuntos
Vesículas Extracelulares/metabolismo , Monócitos/patologia , Infarto do Miocárdio/terapia , Pericárdio/patologia , Animais , Diferenciação Celular , Citocinas/genética , Citocinas/metabolismo , Feminino , Regulação da Expressão Gênica , Contagem de Leucócitos , Ativação Linfocitária/imunologia , Subpopulações de Linfócitos/imunologia , Infarto do Miocárdio/imunologia , Infarto do Miocárdio/patologia , Esferoides Celulares , Suínos
5.
J Vis Exp ; (153)2019 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-31762459

RESUMO

Ischemia-reperfusion injury is the main cause of flap failure in reconstructive microsurgery. The rat is the preferred preclinical animal model in many areas of biomedical research due to its cost-effectiveness and its translation to humans. This protocol describes a method to create a preclinical free skin flap model in rats with ischemia-reperfusion injury. The described 3 cm x 6 cm rat free skin flap model is easily obtained after the placement of several vascular ligatures and the section of the vascular pedicle. Then, 8 h after the ischemic insult and completion of the microsurgical anastomosis, the free skin flap develops the tissue damage. These ischemia-reperfusion injury-related damages can be studied in this model, making it a suitable model for evaluating therapeutic agents to address this pathophysiological process. Furthermore, two main monitoring techniques are described in the protocol for the assessment of this animal model: transit-time ultrasound technology and laser speckle contrast analysis.


Assuntos
Microcirurgia/métodos , Procedimentos de Cirurgia Plástica/métodos , Traumatismo por Reperfusão/etiologia , Animais , Retalhos de Tecido Biológico , Masculino , Ratos , Pele/irrigação sanguínea , Transplante de Pele
6.
Front Pharmacol ; 10: 699, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31316380

RESUMO

Osteoarthritis is one of the most common chronic health conditions associated with pain and disability. Advanced therapies based on mesenchymal stem cells have become valuable options for the treatment of these pathologies. Conditioned serum (CS, "Orthokine") has been used intra-articularly for osteoarthritic patients. In this work, we hypothesized that the rich content on anti-inflammatory proteins and growth factors of CS may exert a beneficial effect on the biological activity of human adipose-derived mesenchymal stem cells (hAdMSCs). In vitro studies were designed using hAdMSCs cocultured with CS at different concentrations (2.5, 5, and 10%). Chondrogenic differentiation assays and immunomodulatory experiments using in vitro-stimulated lymphocytes were performed. Our results demonstrated that CS significantly enhanced the differentiation of hAdMSCs toward chondrocytes. Moreover, hAdMSCs pre-sensitized with CS reduced the lymphocyte proliferation as well as their differentiation toward activated lymphocytes. These results suggest that in vivo coadministration of CS and hAdMSCs may have a beneficial effect on the therapeutic potential of hAdMSCs. Moreover, these results indicate that intra-articular administration of CS might influence the biological behavior of resident stem cells increasing their chondrogenic differentiation and inherent immunomodulatory activity. To our knowledge, this is the first in vitro study reporting this combination.

7.
Stem Cell Res Ther ; 10(1): 152, 2019 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-31151405

RESUMO

BACKGROUND: Allogeneic cardiac-derived progenitor cells (CPC) without immunosuppression could provide an effective ancillary therapy to improve cardiac function in reperfused myocardial infarction. We set out to perform a comprehensive preclinical feasibility and safety evaluation of porcine CPC (pCPC) in the infarcted porcine model, analyzing biodistribution and mid-term efficacy, as well as safety in healthy non-infarcted swine. METHODS: The expression profile of several pCPC isolates was compared with humans using both FACS and RT-qPCR. ELISA was used to compare the functional secretome. One week after infarction, female swine received an intracoronary (IC) infusion of vehicle (CON), 25 × 106 pCPC (25 M), or 50 × 106 pCPC (50 M). Animals were followed up for 10 weeks using serial cardiac magnetic resonance imaging to assess functional and structural remodeling (left ventricular ejection fraction (LVEF), systolic and diastolic volumes, and myocardial salvage index). Statistical comparisons were performed using Kruskal-Wallis and Mann-Whitney U tests. Biodistribution analysis of 18F-FDG-labeled pCPC was also performed 4 h after infarction in a different subset of animals. RESULTS: Phenotypic and functional characterization of pCPC revealed a gene expression profile comparable to their human counterparts as well as preliminary functional equivalence. Left ventricular functional and structural remodeling showed significantly increased LVEF 10 weeks after IC administration of 50 M pCPC, associated to the recovery of left ventricular volumes that returned to pre-infarction values (LVEF at 10 weeks was 42.1 ± 10.0% in CON, 46.5 ± 7.4% in 25 M, and 50.2 ± 4.9% in 50 M, p < 0.05). Infarct remodeling was also improved following pCPC infusion with a significantly higher myocardial salvage index in both treated groups (0.35 ± 0.20 in CON; 0.61 ± 0.20, p = 0.04, in 25 M; and 0.63 ± 0.17, p = 0.01, in 50 M). Biodistribution studies demonstrated cardiac tropism 4 h after IC administration, with substantial myocardial retention of pCPC-associated tracer activity (18% of labeled cells in the heart), and no obstruction of coronary flow, indicating their suitability as a cell therapy product. CONCLUSIONS: IC administration of allogeneic pCPC at 1 week after acute myocardial infarction is feasible, safe, and associated with marked structural and functional benefit. The robust cardiac tropism of pCPC and the paracrine effects on left ventricle post-infarction remodeling established the preclinical bases for the CAREMI clinical trial (NCT02439398).


Assuntos
Miócitos Cardíacos/transplante , Doença Aguda , Animais , Modelos Animais de Doenças , Infarto do Miocárdio , Suínos , Transplante Homólogo
8.
BMC Vet Res ; 15(1): 91, 2019 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-30898123

RESUMO

BACKGROUND: Acute myocardial infarction (AMI) is one of the most deleterious conditions leading to cardiovascular diseases and mortality. The importance of an early and accurate diagnosis assures immediate medical treatments, which are fundamental to reduce mortality and improve prognoses. AMI is associated to an inflammatory response which includes the increase of circulating inflammatory cytokines, chemokines and immune cell activation. This study aimed to identify which are the very early immune-related biomarkers that may be used as predictors of myocardial infarction severity. In order to mimic the pathophysiological events involved in human myocardial infarction, a temporary occlusion (90 min) of the mid-left anterior descending coronary artery was performed in a swine animal model. RESULTS: Lymphocyte subsets analysis in peripheral blood revealed significant alterations in CD4+/CD8+ ratio and naïve and effector/memory T cell percentages at 1 h post-myocardial infarction. Changes in TH1/TH2-related cytokine, monocyte and neutrophil markers gene expression were observed in peripheral blood lymphocytes, as well. Additionally, significant correlations between cardiac parameters (cardiac enzymes, left ventricular ejection fraction and % infarct) and blood-derived parameters (cytokine expression and lymphocyte subset distribution) were found. CONCLUSIONS: Peripheral blood lymphocyte alterations are easily and swiftly detectable, so they may be good biomarkers for a very early prognosis and to predict myocardial infarction severity.


Assuntos
Inflamação/veterinária , Infarto do Miocárdio/veterinária , Doenças dos Suínos/diagnóstico , Animais , Biomarcadores/sangue , Relação CD4-CD8/veterinária , Creatina Quinase Forma MB/sangue , Citocinas/sangue , Modelos Animais de Doenças , Feminino , Citometria de Fluxo/veterinária , Testes de Função Cardíaca , Inflamação/sangue , Inflamação/diagnóstico , Subpopulações de Linfócitos , Linfócitos/metabolismo , Infarto do Miocárdio/sangue , Infarto do Miocárdio/diagnóstico , Suínos , Doenças dos Suínos/sangue , Troponina I/sangue
9.
Biol Reprod ; 100(5): 1180-1192, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30596891

RESUMO

Advanced age is a risk factor undermining women's fertility. Hence, the optimization of assisted reproduction techniques is an interdisciplinary challenge that requires the improvement of in vitro culture systems. Here, we hypothesize that supplementation of embryo culture medium with extracellular vesicles from endometrial-derived mesenchymal stem cells (EV-endMSCs) may have a positive impact on the embryo competence of aged oocytes. In this work, 24 weeks old B6D2 female mice were used as egg donors and in vitro fertilization assays were performed using males from the same strain (8-12 weeks); the presumptive zygotes were incubated in the presence of 0, 10, 20, 40, or 80 µg/ml of EV-endMSCs. The results from the proteomic analysis of EV-endMSCs and the classification by Reactome pathways allowed us to identify proteins closely related with the fertilization process. Moreover, in our aged murine model, the supplementation of the embryo culture medium with EV-endMSCs improved the developmental competence of the embryos as well as the total blastomere count. Finally, gene expression analysis of murine blastocysts showed significant changes on core genes related to cellular response to oxidative stress, metabolism, placentation, and trophectoderm/inner cell mass formation. In summary, we demonstrate that EV-endMSCs increase the quality of the embryos, and according to proteomic and genomic analysis, presumably by modulating the expression of antioxidant enzymes and promoting pluripotent activity. Therefore, EV-endMSCs could be a valuable tool in human assisted reproduction improving the developmental competence of aged oocytes and increasing the odds of implantation and subsequent delivery.


Assuntos
Senescência Celular/fisiologia , Embrião de Mamíferos , Endométrio/citologia , Vesículas Extracelulares/fisiologia , Idade Materna , Células-Tronco Mesenquimais/ultraestrutura , Recuperação de Oócitos , Animais , Células Cultivadas , Técnicas de Cocultura/métodos , Técnicas de Cocultura/normas , Técnicas de Cocultura/veterinária , Técnicas de Cultura Embrionária/normas , Técnicas de Cultura Embrionária/veterinária , Feminino , Fertilização in vitro/normas , Fertilização in vitro/veterinária , Humanos , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Recuperação de Oócitos/métodos , Recuperação de Oócitos/normas , Recuperação de Oócitos/veterinária , Oócitos/citologia , Oócitos/fisiologia , Controle de Qualidade
10.
Artigo em Inglês | MEDLINE | ID: mdl-31921832

RESUMO

Endometrial-derived Mesenchymal Stem Cells (endMSCs) are involved in the regeneration and remodeling of human endometrium, being considered one of the most promising candidates for stem cell-based therapies. Their therapeutic effects have been found to be mediated by extracellular vesicles (EV-endMSCs) with pro-angiogenic, anti-apoptotic, and immunomodulatory effects. Based on that, the main goal of this study was to characterize the proteome and microRNAome of these EV-endMSCs by proteomics and transcriptomics approaches. Additionally, we hypothesized that inflammatory priming of endMSCs may contribute to modify the therapeutic potential of these vesicles. High-throughput proteomics revealed that 617 proteins were functionally annotated as Extracellular exosome (GO:0070062), corresponding to the 70% of the EV-endMSC proteome. Bioinformatics analyses allowed us to identify that these proteins were involved in adaptive/innate immune response, complement activation, antigen processing/presentation, negative regulation of apoptosis, and different signaling pathways, among others. Of note, multiplexed quantitative proteomics and Systems Biology analyses showed that IFNγ priming significantly modulated the protein profile of these vesicles. As expected, proteins involved in antigen processing and presentation were significantly increased. Interestingly, immunomodulatory proteins, such as CSF1, ERAP1, or PYCARD were modified. Regarding miRNAs expression profile in EV-endMSCs, Next-Generation Sequencing (NGS) showed that the preferred site of microRNAome targeting was the nucleus (n = 371 microTargets), significantly affecting signal transduction (GO:0007165), cell proliferation (GO:0008283), and apoptotic processes (GO:0006915), among others. Interestingly, NGS analyses highlighted that several miRNAs, such as hsa-miR-150-5p or hsa-miR-196b-5p, were differentially expressed in IFNγ-primed EV-endMSCs. These miRNAs have a functional involvement in glucocorticoid receptor signaling, IL-6/8/12 signaling, and in the role of macrophages. In summary, these results allowed us to understand the complexity of the molecular networks in EV-endMSCs and their potential effects on target cells. To our knowledge, this is the first comprehensive study based on proteomic and genomic approaches to unravel the therapeutic potential of these extracellular vesicles, that may be used as immunomodulatory effectors in the treatment of inflammatory conditions.

11.
PLoS One ; 13(12): e0209624, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30589864

RESUMO

Ischemia reperfusion injury is associated with tissue damage and inflammation, and is one of the main factors causing flap failure in reconstructive microsurgery. Although ischemia-reperfusion (I/R) injury is a well-studied aspect of flap survival, its biological mechanisms remain to be elucidated. To better understand the biological processes of ischemia reperfusion injury, and to develop further therapeutic strategies, the main objective of this study was to identify the gene expression pattern and histological changes in an I/R injury animal model. Fourteen rats (n = 7/group) were randomly divided into control or ischemia-reperfusion group (8 hours of ischemia). Microsurgical anastomoses were objectively assessed using transit-time-ultrasound technology. Seven days after surgery, flap survival was evaluated and tissue samples were harvested for anatomopathological and gene-expression analyses.The I/R injury reduced the survival of free flaps and histological analyses revealed a subcutaneous edema together with an inflammatory infiltrate. Interestingly, the Arginase 1 expression level as well as the ratio of Arginase 1/Nitric oxide synthase 2 showed a significant increase in the I/R group. In summary, here we describe a well-characterized I/R animal model that may serve to evaluate therapeutic agents under reproducible and controlled conditions. Moreover, this model could be especially useful for the evaluation of arginase inhibitors and different compounds of potential interest in reconstructive microsurgery.


Assuntos
Retalhos de Tecido Biológico/irrigação sanguínea , Microvasos , Traumatismo por Reperfusão/etiologia , Traumatismo por Reperfusão/patologia , Animais , Biomarcadores , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Sobrevivência de Enxerto , Imuno-Histoquímica , Masculino , Microscopia , Ratos , Transplante de Pele , Ultrassonografia
12.
Vet Immunol Immunopathol ; 205: 49-57, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30459001

RESUMO

Preclinical studies in cardiovascular medicine are necessary to translate basic research to the clinic. The porcine model has been widely used to understand the biological mechanisms involved in cardiovascular disorders for which purpose different closed-chest models have been developed in the last years to mimic the pathophysiological events seen in human myocardial infarction. In this work, we studied hematological, biochemical and immunological parameters, as well as Magnetic resonance derived cardiac function measurements obtained from a swine myocardial infarction model. We identified some blood parameters which were significantly altered after myocardial infarction induction. More importantly, these parameters (gamma-glutamyl transferase, glutamic pyruvic transaminase, red blood cell counts, hemoglobin concentration, hematocrit, platelet count and plateletcrit) correlated positively with cardiac function, infarct size and/or cardiac enzymes (troponin I and creatine kinase-MB). Thus several blood-derived parameters have allowed us to predict the severity of myocardial infarction in a clinically relevant animal model. Therefore, here we provide a simple, affordable and reliable way that could prove useful in the follow up of myocardial infarction and in the evaluation of new therapeutic strategies in this animal model.


Assuntos
Biomarcadores/sangue , Infarto do Miocárdio/sangue , Infarto do Miocárdio/imunologia , Alanina Transaminase/sangue , Animais , Modelos Animais de Doenças , Contagem de Eritrócitos , Feminino , Testes de Função Cardíaca , Hematócrito , Hemoglobinas/análise , Imageamento por Ressonância Magnética , Masculino , Infarto do Miocárdio/diagnóstico por imagem , Estudos Prospectivos , Suínos , gama-Glutamiltransferase/sangue
13.
Reprod Domest Anim ; 53 Suppl 2: 46-49, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30238659

RESUMO

Advanced age reduces the success of in vitro fertilization (IVF) being this effect partly mediated by an overproduction of reactive oxygen species (ROS) that trigger apoptosis. It has been demonstrated that extracellular vesicles derived from endometrial mesenchymal stem cells (EV-endMSCs) exert an antioxidant effect and can be used as IVF coadjutants. In this work, endMSCs were isolated from human menstrual blood (n = 4) and characterized according to multipotentiality and surface marker expression prior EV-endMSCs isolation. Oocytes were obtained from 21 B6D2 mice (24 weeks) and coincubated with sperm from young males (8-12 weeks). Presumptive zygotes were incubated in the presence of 0, 10, 20, 40 or 80 µg/ml of EV-endMSCs in KSOM medium. Blastocyst yield was evaluated, and 25 blastocysts per group were used for qPCR. Blastocyst rate was 29.4% in control; 45.2% for 10 µg/ml, 62.9% for 20 µg/ml, 55.5% for 40 µg/ml and 53.8% in the 80 µg/ml (n = 124-130 oocytes) being all the increases significantly different when compared against control (p < 0.05). The 20-80 µg/ml treatments decreased the expression of glutathione peroxidase (Gpx1), and the 10-40 µg/ml treatments reduced the expression of superoxide dismutase (Sod1; p < 0.05) compared to control; Bax mRNA expression did not vary. Our results suggest that the increased developmental competence of the embryos could be partly mediated by the EV-endMSCs' ROS scavenger activity.


Assuntos
Blastocisto/fisiologia , Endométrio/fisiologia , Vesículas Extracelulares/fisiologia , Fertilização in vitro/veterinária , Células-Tronco Mesenquimais/citologia , Animais , Modelos Animais de Doenças , Desenvolvimento Embrionário , Feminino , Expressão Gênica , Humanos , Técnicas de Maturação in Vitro de Oócitos/veterinária , Masculino , Camundongos , Espécies Reativas de Oxigênio/metabolismo , Espermatozoides , Zigoto
14.
J Tissue Eng Regen Med ; 12(10): 2088-2098, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30058282

RESUMO

Endometrial mesenchymal stem cells (endMSCs) reside in the basal and functional layer of human endometrium and participate in tissue remodelling, which is required for maintaining the regenerative capacity of the endometrium. The endMSCs are multipotent stem cells and exhibit immunomodulatory effects. This paper aimed to evaluate the regulatory effects of extracellular vesicles derived from endMSCs (EV-endMSCs) in the setting of T cell activation. In vitro stimulations of lymphocytes were performed in the presence of EV-endMSCs. These in vitro-stimulated lymphocytes were functionally and phenotypically characterized to distinguish CD4+ and CD8+ T cell differentiation subsets. Moreover, the inhibition of TGFß was performed with neutralizing antibodies. The phenotype and nanoparticle tracking analysis of the EV-endMSCs demonstrated that they are similar in terms of size distribution to other mesenchymal stem cells-derived exosomes. The in vitro assays showed an immunomodulatory potential of these vesicles to counteract the differentiation of CD4+ T cells. The quantification of active TGFß in EV-endMSCs was found to be very high when compared with extracellular vesicles-free concentrated supernatants. Finally, the neutralization of TGFß significantly attenuated the immunomodulatory activity of EV-endMSCs. In summary, this is the first report demonstrating that EV-endMSCs exhibit a potent inhibitory effect against CD4+ T cell activation, which is partially mediated by TGFß signalling.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Endométrio/citologia , Vesículas Extracelulares/metabolismo , Imunomodulação , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Técnicas de Cocultura , Feminino , Humanos , Fenótipo
15.
J Reconstr Microsurg ; 34(8): 601-609, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29723883

RESUMO

BACKGROUND: Ischemia-reperfusion (I/R) injury is inevitable during free tissue transfers. When the period of ischemia exceeds the tissue tolerance, it causes necrosis and flap failure. The aim of this study was to investigate the effects of adipose-derived stem cells (ASCs) embedded in a collagen type I scaffold on the survival of free skin flaps to counteract I/R injury. METHODS: Left superficial caudal epigastric skin flaps (3 × 6 cm) were performed in 28 Wistar rats that were divided into four groups. The flaps elevated in the animals of the control group did not suffer any ischemic insult, and the vascular pedicle was not cut. All other flaps were subjected to 8 hours of ischemia prior to revascularization: I/R control group (8 hours of ischemia), I/R scaffold group (8 hours of ischemia + collagen type I scaffold), and I/R scaffold-ASCs group (8 hours of ischemia + collagen type I scaffold with rat ASCs embedded). Transit-time ultrasound blood flow measurements were performed. After 7 days, the areas of flap survival were measured and tissues were stained with hematoxylin/eosin and Masson's trichrome stain for histological analysis. RESULTS: The mean percentage flap survival area was significantly higher in the ASCs-treated flaps (I/R scaffold-ASCs group) compared with the ischemic controls (I/R control group and I/R scaffold group). Higher vascular proliferation and lower severity of necrosis and inflammatory changes were seen histologically in the samples of the ASCs-treated group. No significant difference in blood flow was detected between groups. CONCLUSION: Subcutaneous administration of ASCs embedded on a collagen type I scaffold reduces tissue damage after I/R injury in microvascular free flaps.


Assuntos
Tecido Adiposo/citologia , Retalhos de Tecido Biológico , Traumatismo por Reperfusão/patologia , Pele/irrigação sanguínea , Transplante de Células-Tronco/métodos , Animais , Modelos Animais de Doenças , Sobrevivência de Enxerto , Masculino , Ratos , Ratos Wistar , Traumatismo por Reperfusão/cirurgia
16.
PLoS One ; 13(4): e0196080, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29684038

RESUMO

Endometrial Mesenchymal Stromal Cells (endMSCs) are multipotent cells with immunomodulatory and pro-regenerative activity which is mainly mediated by a paracrine effect. The exosomes released by MSCs have become a promising therapeutic tool for the treatment of immune-mediated diseases. More specifically, extracellular vesicles derived from endMSCs (EV-endMSCs) have demonstrated a cardioprotective effect through the release of anti-apoptotic and pro-angiogenic factors. Here we hypothesize that EV-endMSCs may be used as a co-adjuvant to improve in vitro fertilization outcomes and embryo quality. Firstly, endMSCs and EV-endMSCs were isolated and phenotypically characterized for in vitro assays. Then, in vitro studies were performed on murine embryos co-cultured with EV-endMSCs at different concentrations. Our results firstly demonstrated a significant increase on the total blastomere count of expanded murine blastocysts. Moreover, EV-endMSCs triggered the release of pro-angiogenic molecules from embryos demonstrating an EV-endMSCs concentration-dependent increase of VEGF and PDGF-AA. The release of VEGF and PDGF-AA by the embryos may indicate that the beneficial effect of EV-endMSCs could be mediating not only an increase in the blastocyst's total cell number, but also may promote endometrial angiogenesis, vascularization, differentiation and tissue remodeling. In summary, these results could be relevant for assisted reproduction being the first report describing the beneficial effect of human EV-endMSCs on embryo development.


Assuntos
Blastômeros/citologia , Endométrio/citologia , Vesículas Extracelulares/fisiologia , Fator de Crescimento Derivado de Plaquetas/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Blastômeros/fisiologia , Diferenciação Celular , Técnicas de Cocultura , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário , Endométrio/metabolismo , Feminino , Fertilização in vitro , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo
17.
An Real Acad Farm ; 84(1): 39-51, ene.-mar. 2018. ilus, tab, graf
Artigo em Espanhol | IBECS | ID: ibc-178048

RESUMO

Una hernia incisional consiste en una protrusión de tejido a través de una cicatriz traumática o quirúrgica en la pared abdominal. El tratamiento de este y de otros tipos de hernias pasa frecuentemente por la implantación de una malla quirúrgica para reforzar el tejido debilitado. Sin embargo, a menudo se produce una respuesta inflamatoria exacerbada que desemboca en diferentes complicaciones, teniendo consecuencias graves para el paciente. Considerando el potencial inmunomodulador de las células madre mesenquimales (MSCs) y de sus exosomas (exo-MSCs), en este estudio planteamos que la administración de ambos productos terapéuticos, conjuntamente con los selladores de fibrina que se utilizan frecuentemente para la fijación de las mallas quirúrgicas, podría ejercer un efecto biológico y terapéutico que ayudara a controlar esa respuesta inflamatoria y mejorara, por tanto, el éxito del tratamiento con mallas quirúrgicas. Los resultados obtenidos en este estudio mostraron, en un modelo murino de hernia incisional, que los exo-MSCs reducen la infiltración de macrófagos inflamatorios M1 y que existe una predominancia de citoquinas relacionadas con la respuesta Th2, y con ello, con la polarización de macrófagos hacia un fenotipo M2 antiinflamatorio, en el tejido circundante a las mallas en las que se vehicularon MSCs o sus exosomas. Este estudio concluye que la fijación de mallas quirúrgicas con selladores de fibrina combinados con MSCs o exo-MSCs tendría un efecto beneficioso en el tratamiento de la hernia incisional, en términos de reducción de la respuesta inflamatoria y modulación de una reacción exacerbada frente a un cuerpo extraño


An incisional hernia constitutes a tissue protrusion through a traumatic or surgical scar in the abdominal wall. Frequently, the treatment of the incisional hernia, as well as other types of hernia, involves the implantation of a surgical mesh to reinforce the weakened tissue. However, an exacerbated inflammatory response is commonly developed after this implantation, having serious consequences for the patient. Considering the immunomodulatory potential of mesenchymal stem cells (MSCs) and their exosomes (exo-MSCs), in this study we proposed that the administration of these two therapeutic products, together with fibrin sealants that are frequently used to fix surgical meshes, could have a beneficial biological and therapeutic effect that could help to modulate the inflammatory response and improve the success of the surgical mesh implantation. The results obtained in this work showed, in a murine model of incisional hernia, that exo-MSCs reduce M1 inflammatory macrophages infiltration and that there is a predominance of Th2- related cytokines in the surrounding tissue of MSCs or exo-MSCs treated meshes, favoring the macrophage polarization towards a M2 anti-inflammatory phenotype. This study concludes that mesh fixation with fibrin sealants co-administered with MSCs or exo-MSCs would have a beneficiary effect on the treatment of incisional hernia in terms of reduction of the inflammatory response and modulation of the foreign body reaction


Assuntos
Animais , Camundongos , Células-Tronco , Exossomos , Hérnia/tratamento farmacológico , Telas Cirúrgicas , Proteínas Inflamatórias de Macrófagos/uso terapêutico , Medula Óssea , Citometria de Fluxo/métodos , Citometria de Fluxo/veterinária
18.
Acta Biomater ; 71: 318-329, 2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29462710

RESUMO

Surgical meshes are effective and frequently used to reinforce soft tissues. Fibrin glue (FG) has been widely used for mesh fixation and is also considered an optimal vehicle for stem cell delivery. The aim of this preclinical study was to evaluate the therapeutic effect of MSCs and their exosomes combined with FG for the treatment of incisional hernia. A murine incisional hernia model was used to implant surgical meshes and different treatments with FG, MSCs and exo-MSCs were applied. The implanted meshes were evaluated at day 7 by anatomopathology, cellular analysis of infiltrating leukocytes and gene expression analysis of TH1/TH2 cytokines, MMPs, TIMPs and collagens. Our results demonstrated a significant increase of anti-inflammatory M2 macrophages and TH2 cytokines when MSCs or exo-MSCs were used. Moreover, the analysis of MMPs, TIMPs and collagen exerted significant differences in the extracellular matrix and in the remodeling process. Our in vivo study suggests that the fixation of surgical meshes with FG and MSCs or exo-MSCs will have a beneficial effect for the treatment of incisional hernia in terms of improved outcomes of damaged tissue, and especially, in the modulation of inflammatory responses towards a less aggressive and pro-regenerative profile. STATEMENT OF SIGNIFICANCE: The implantation of surgical meshes is the standard procedure to reinforce tissue defects such as hernias. However, an exacerbated and persistent inflammatory response secondary to this implantation is frequently observed, leading to a strong discomfort and chronic pain in the patients. In many cases, an additional surgical intervention is needed to remove the mesh. This study shows that mesenchymal stem cells and their exosomes, combined with a fibrin sealant, can be used for the successful fixation of these meshes. This new therapeutic approach, assayed in a murine model of incisional hernia, favors the modulation of the inflammatory response towards a less aggressive and pro-regenerative profile.


Assuntos
Exossomos/imunologia , Adesivo Tecidual de Fibrina/farmacologia , Hérnia Abdominal , Herniorrafia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/imunologia , Animais , Citocinas/imunologia , Modelos Animais de Doenças , Exossomos/patologia , Hérnia Abdominal/imunologia , Hérnia Abdominal/patologia , Hérnia Abdominal/terapia , Inflamação/imunologia , Inflamação/patologia , Inflamação/terapia , Macrófagos/imunologia , Macrófagos/patologia , Células-Tronco Mesenquimais/patologia , Camundongos , Camundongos Endogâmicos ICR , Células Th1/imunologia , Células Th1/patologia , Células Th2/imunologia , Células Th2/patologia
19.
Front Vet Sci ; 4: 39, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28377922

RESUMO

Synovitis is an inflammatory process associated with pain, disability, and discomfort, which is usually treated with anti-inflammatory drugs or biological agents. Mesenchymal stem cells (MSCs) have been also successfully used in the treatment of inflammatory-related diseases such as synovitis or arthritis. In the last years, the exosomes derived from MSCs have become a promising tool for the treatment of inflammatory-related diseases and their therapeutic effect is thought to be mediated (at least in part) by their immunomodulatory potential. In this work, we aimed to evaluate the anti-inflammatory effect of these exosomes in an antigen-induced synovitis animal model. To our knowledge, this is the first report where exosomes derived from MSCs have been evaluated in an animal model of synovitis. Our results demonstrated a decrease of synovial lymphocytes together with a downregulation of TNF-α transcripts in those exosome-treated joints. These results support the immunomodulatory effect of these exosomes and point out that they may represent a promising therapeutic option for the treatment of synovitis.

20.
BMC Vet Res ; 13(1): 93, 2017 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-28388908

RESUMO

BACKGROUND: Synovitis is an inflammation-related disease linked to rheumatoid arthritis, osteoarthritis, infections and trauma. This inflammation is accompanied by immune cells infiltration which initiates an inflammatory response causing pain, discomfort and affecting the normal joint function. The treatment of synovitis is based on the administration of anti-inflammatory drugs or biological agents such as platelet rich plasma and mesenchymal stem cells. However, the evaluation and validation of more effective therapies of synovitis requires the establishment of clinically relevant animal models. RESULTS: In this study, Large White pigs were pre-immunized to evaluate an antigen-induced synovitis. The immune monitoring of synovial fluids in this model allowed us the identification of IL-12p40 and T cell subsets as immune biomarkers. Moreover, the evolution of synovitis was performed by arthroscopic procedures and kinetic analysis. In summary, this paper describes an animal model of antigen-induced synovitis to be used in the evaluation of anti-inflammatory therapies. CONCLUSIONS: The novelty of this paper lies in the development of a clinically relevant model of synovitis which permits the simultaneous evaluation of synovitis from an immunological, surgical and kinetic point of view.


Assuntos
Articulações do Carpo , Modelos Animais de Doenças , Inflamação/veterinária , Soroalbumina Bovina/imunologia , Sus scrofa , Sinovite/veterinária , Animais , Artroscopia/veterinária , Biomarcadores , Inflamação/induzido quimicamente , Subunidade p40 da Interleucina-12/metabolismo , Suínos , Doenças dos Suínos/etiologia , Líquido Sinovial/citologia , Sinovite/induzido quimicamente , Sinovite/imunologia , Subpopulações de Linfócitos T/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...