2
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Inhibiting DNA methylation as a strategy to enhance adipose-derived stem cells differentiation: Focus on the role of Akt/mTOR and Wnt/β-catenin pathways on adipogenesis

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Adipose-derived mesenchymal stem cells (ASCs) represent a valid therapeutic option for clinical application in several diseases, due to their ability to repair damaged tissues and to mitigate the inflammatory/immune response. A better understanding of the underlying mechanisms regulating ASC biology might represent the chance to modulate their in vitro characteristics and differentiation potential for regenerative medicine purposes. Herein, we investigated the effects of the demethylating agent 5-azacytidine (5-aza) on proliferation, clonogenicity, migration, adipogenic differentiation and senescence of ASCs, to identify the molecular pathways involved. Through functional assays, we observed a detrimental effect of 5-aza on ASC self-renewal capacity and migration, accompanied by actin cytoskeleton reorganization, with decreased stress fibers. Conversely, 5-aza treatment enhanced ASC adipogenic differentiation, as assessed by lipid accumulation and expression of lineage-specific markers. We analyzed the involvement of the Akt/mTOR, MAPK and Wnt/β-catenin pathways in these processes. Our results indicated impairment of Akt and ERK phosphorylation, potentially explaining the reduced cell proliferation and migration. We observed a 5-aza-mediated inhibition of the Wnt signaling pathway, this potentially explaining the pro-adipogenic effect of the drug. Finally, 5-aza treatment significantly induced ASC senescence, through upregulation of the p53/p21 axis. Our data may have important translational implications, by helping in clarifying the potential risks and advantages of using epigenetic treatment to improve ASC characteristics for cell-based clinical approaches.

          Related collections

          Most cited references77

          • Record: found
          • Abstract: found
          • Article: not found

          The senescence-associated secretory phenotype: the dark side of tumor suppression.

          Cellular senescence is a tumor-suppressive mechanism that permanently arrests cells at risk for malignant transformation. However, accumulating evidence shows that senescent cells can have deleterious effects on the tissue microenvironment. The most significant of these effects is the acquisition of a senescence-associated secretory phenotype (SASP) that turns senescent fibroblasts into proinflammatory cells that have the ability to promote tumor progression.
            Bookmark
            • Record: found
            • Abstract: not found
            • Article: not found

            Hallmarks of Cellular Senescence

              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Human adipose tissue is a source of multipotent stem cells.

              Much of the work conducted on adult stem cells has focused on mesenchymal stem cells (MSCs) found within the bone marrow stroma. Adipose tissue, like bone marrow, is derived from the embryonic mesenchyme and contains a stroma that is easily isolated. Preliminary studies have recently identified a putative stem cell population within the adipose stromal compartment. This cell population, termed processed lipoaspirate (PLA) cells, can be isolated from human lipoaspirates and, like MSCs, differentiate toward the osteogenic, adipogenic, myogenic, and chondrogenic lineages. To confirm whether adipose tissue contains stem cells, the PLA population and multiple clonal isolates were analyzed using several molecular and biochemical approaches. PLA cells expressed multiple CD marker antigens similar to those observed on MSCs. Mesodermal lineage induction of PLA cells and clones resulted in the expression of multiple lineage-specific genes and proteins. Furthermore, biochemical analysis also confirmed lineage-specific activity. In addition to mesodermal capacity, PLA cells and clones differentiated into putative neurogenic cells, exhibiting a neuronal-like morphology and expressing several proteins consistent with the neuronal phenotype. Finally, PLA cells exhibited unique characteristics distinct from those seen in MSCs, including differences in CD marker profile and gene expression.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Cell Dev Biol
                Front Cell Dev Biol
                Front. Cell Dev. Biol.
                Frontiers in Cell and Developmental Biology
                Frontiers Media S.A.
                2296-634X
                02 September 2022
                2022
                : 10
                : 926180
                Affiliations
                [1] 1 Department of Experimental Medicine , Sapienza University of Rome , Rome, Italy
                [2] 2 Department of Maternal, Infantile and Urological Sciences , Sapienza University of Rome , Rome, Italy
                [3] 3 Department of Sense Organs , Sapienza University of Rome , Rome, Italy
                [4] 4 Department of Surgery “P. Valdoni” , Unit of Plastic Surgery “P. Valdoni” , Sapienza University of Rome , Rome, Italy
                [5] 5 Department of Advanced Medical and Surgical Sciences (DAMSS) , University of Campania “Luigi Vanvitelli” , Naples, Italy
                Author notes

                Edited by: Lindolfo da Silva Meirelles, Universidade Luterana do Brasil, Brazil

                Reviewed by: Borhane Guezguez, German Cancer Research Center (DKFZ), Germany

                Meifeng Xu, University of Cincinnati, United States

                *Correspondence: S. Ceccarelli , simona.ceccarelli@ 123456uniroma1.it

                This article was submitted to Stem Cell Research, a section of the journal Frontiers in Cell and Developmental Biology

                Article
                926180
                10.3389/fcell.2022.926180
                9478209
                678a228e-df4d-4203-b29f-adaf866eb707
                Copyright © 2022 Ceccarelli, Gerini, Megiorni, Pontecorvi, Messina, Camero, Anastasiadou, Romano, Onesti, Napoli and Marchese.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 22 April 2022
                : 28 July 2022
                Funding
                Funded by: Ministero dell'Università e della Ricerca , doi 10.13039/501100021856;
                Categories
                Cell and Developmental Biology
                Original Research

                adipose-derived adult stem cells (ascs),epigenetic regulation,5-azacytidine,adipogenesis,akt/mtor,wnt/ beta-catenin pathway

                Comments

                Comment on this article