4 research outputs found

    Role of mesenchymal stem cells in the remodeling of the extracellular matrix in glioblastoma

    Get PDF
    Motivation: Glioblastoma multiforme (GBM) is the most common and malignant brain tumor. Patients who suffer from this malignancy have a median survival rate of approximately 12 months. Current treatments for GBM are surgery, radiotherapy, and chemotherapy (1), but these treatments do not offer a definitive solution. In this context, the cell therapy based on mesenchymal stem cells (MSCs) has emerged as a promising alternative to improve the survival of patients (2). Our study aims to investigate the underlying mechanisms by which MSCs could exert beneficial effects in GBM, paying special attention to the extracellular matrix remodeling. Methods: All in vitro and in vivo studies were performed using three experimental groups: U87 glioma cells (U87), U87 glioma cells in the presence of MSCs (U87+MSC), and U87 glioma cells in the presence of MSCs previously pretreated with a substance that improves their therapeutic properties (U87+MSCpret). First, using paraffin-embedding tissuse sections of tumors that were subcutaneously grown in immunosuppressed mice (n=5), we performed a Sirius Red staining to quantify the collagen fibers content. Second, using the transwell coculture system (3),  we conducted an RNA-Seq analysis to evaluate the expression levels of genes involved in extracellular matrix remodeling in the U87 glioma cells of each experimental group (n=3). RT-qPCR was performed to validate the RNA-Seq results (n=3). Values of p <0.05 were considered statistically significant. Results: The quantification of the collagen deposition in subcutaneous tumors showed a tendency to decrease in the U87+MSCpret group. In the RNA-Seq analysis, we obtained a list of extracellular matrix-related genes with a significant differential expression (e.g. Mmp28, Hyal1, Col24a1, Cemip, Eln). RT-qPCR results showed a tendency to reduce the expression of genes related to the remodeling of the extracellular matrix, such as Mmp9, Mmp2, Timp1, Timp2 or Tgf-β, in the U87+MSCpret group. Of note, this reduction in the expression levels became significant for Timp1 (p<0.05). Conclusions: Pretreated MSCs prevent the accumulation of collagen in the GMB using in vivo models, which is associated with less invasive tumors. In vitro, we identified that the expression of extracellular matrix proteins tended to decrease in GBM cells when they were co-cultured with pretreated MSCs, suggesting that these cells may reduce tumorigenesis by altering extracellular matrix remodeling

    Metabolic reprogramming by Acly inhibition using SB-204990 alters glucoregulation and modulates molecular mechanisms associated with aging

    Get PDF
    19 Páginas.-- 7 FigurasATP-citrate lyase is a central integrator of cellular metabolism in the interface of protein, carbohydrate, and lipid metabolism. The physiological consequences as well as the molecular mechanisms orchestrating the response to long-term pharmacologically induced Acly inhibition are unknown. We report here that the Acly inhibitor SB-204990 improves metabolic health and physical strength in wild-type mice when fed with a high-fat diet, while in mice fed with healthy diet results in metabolic imbalance and moderated insulin resistance. By applying a multiomic approach using untargeted metabolomics, transcriptomics, and proteomics, we determined that, in vivo, SB-204990 plays a role in the regulation of molecular mechanisms associated with aging, such as energy metabolism, mitochondrial function, mTOR signaling, and folate cycle, while global alterations on histone acetylation are absent. Our findings indicate a mechanism for regulating molecular pathways of aging that prevents the development of metabolic abnormalities associated with unhealthy dieting. This strategy might be explored for devising therapeutic approaches to prevent metabolic diseases.This work was funded by grants from the Ministerio de Economía y Competitividad, Instituto de Salud Carlos III, co-funded by Fondos FEDER (PI15/00134, PI18/01590, CPII19/00023 to A.M.M.) and the Ministerio de Ciencia e Innovación (PID2021-123965OB-100 to A.M.M.). A.M.M. is funded by the Junta de Andalucía P20_00480, the Spanish Society of Diabetes, and CSIC 202220I059. M.S.K. is funded by the Nordea Foundation (#02-2017-1749), the Novo Nordisk Foundation (#NNF17OC0027812), the Neye Foundation, the Lundbeck Foundation (#R324-2019-1492), the Ministry of Higher Education and Science of Denmark (#0238-00003B). V.C.G. is funded by the Instituto de Salud Carlos III (CP19/00046), co-funded by FEDER. F.M. is funded by the CIBERDEM of the Instituto de Salud Carlos III. A.M.M. is the guarantor of this work and, as such, had full access to all the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis. We acknowledge the support of the group of basic research on diabetes of the Spanish Society of Diabetes.Peer reviewe

    Preclinical Safety Evaluation of Intranasally Delivered Human Mesenchymal Stem Cells in Juvenile Mice

    No full text
    Mesenchymal stem cell (MSC)-based therapy is a promising therapeutic approach in the management of several pathologies, including central nervous system diseases. Previously, we demonstrated the therapeutic potential of human adipose-derived MSCs for neurological sequelae of oncological radiotherapy using the intranasal route as a non-invasive delivery method. However, a comprehensive investigation of the safety of intranasal MSC treatment should be performed before clinical applications. Here, we cultured human MSCs in compliance with quality control standards and administrated repeated doses of cells into the nostrils of juvenile immunodeficient mice, mimicking the design of a subsequent clinical trial. Short- and long-term effects of cell administration were evaluated by in vivo and ex vivo studies. No serious adverse events were reported on mouse welfare, behavioral performances, and blood plasma analysis. Magnetic resonance study and histological analysis did not reveal tumor formation or other abnormalities in the examined organs of mice receiving MSCs. Biodistribution study reveals a progressive disappearance of transplanted cells that was further supported by an absent expression of human GAPDH gene in the major organs of transplanted mice. Our data indicate that the intranasal application of MSCs is a safe, simple and non-invasive strategy and encourage its use in future clinical trials.This work was funded by grants from the Andalusian Regional Ministry of Health (PI 0272-2017 to V.C.-G), the Institute of Health Carlos III co-funded by Fondos FEDER (PI20/00341, CP19/00046 to V.C.-G; PI18/01590, CPII19/00023 to A.M.-M.), the Fundación Científica de la Aso ciación Española Contra el Cáncer (IDEAS20051CAPI to V.C.-G.), the crowdfunding platform PRE CIPITA of the Spanish Foundation for Science and Technology (2018-000237 to V.C.-G.), and the Asociación Pablo Ugarte (+ VIDA project to V.C.-G).Ye
    corecore