Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Mickael Deschepper is active.

Publication


Featured researches published by Mickael Deschepper.


Stem Cells Translational Medicine | 2015

Oxygen Tension Regulates Human Mesenchymal Stem Cell Paracrine Functions

Joseph Paquet; Mickael Deschepper; Adrien Moya; Delphine Logeart-Avramoglou; Catherine Boisson-Vidal; Hervé Petite

Mesenchymal stem cells (MSCs) have captured the attention and research endeavors of the scientific world because of their differentiation potential. However, there is accumulating evidence suggesting that the beneficial effects of MSCs are predominantly due to the multitude of bioactive mediators secreted by these cells. Because the paracrine potential of MSCs is closely related to their microenvironment, the present study investigated and characterized select aspects of the human MSC (hMSC) secretome and assessed its in vitro and in vivo bioactivity as a function of oxygen tension, specifically near anoxia (0.1% O2) and hypoxia (5% O2), conditions that reflect the environment to which MSCs are exposed during MSC‐based therapies in vivo. In contrast to supernatant conditioned media (CM) obtained from hMSCs cultured at either 5% or 21% of O2, CM from hMSCs cultured under near anoxia exhibited significantly (p < .05) enhanced chemotactic and proangiogenic properties and a significant (p < .05) decrease in the inflammatory mediator content. An analysis of the hMSC secretome revealed a specific profile under near anoxia: hMSCs increase their paracrine expression of the angiogenic mediators vascular endothelial growth factor (VEGF)‐A, VEGF‐C, interleukin‐8, RANTES, and monocyte chemoattractant protein 1 but significantly decrease expression of several inflammatory/immunomodulatory mediators. These findings provide new evidence that elucidates aspects of great importance for the use of MSCs in regenerative medicine and could contribute to improving the efficacy of such therapies.


Stem Cells | 2013

Proangiogenic and Prosurvival Functions of Glucose in Human Mesenchymal Stem Cells upon Transplantation

Mickael Deschepper; Mathieu Manassero; Karim Oudina; Joseph Paquet; Laurent‐Emmanuel Monfoulet; Morad Bensidhoum; Delphine Logeart-Avramoglou; Hervé Petite

A major limitation in the development of cellular therapies using human mesenchymal stem cells (hMSCs) is cell survival post‐transplantation. In this study, we challenged the current paradigm of hMSC survival, which assigned a pivotal role to oxygen, by testing the hypothesis that exogenous glucose may be key to hMSC survival. We demonstrated that hMSCs could endure sustained near‐anoxia conditions only in the presence of glucose. In this in vitro cell model, the protein expressions of Hif‐1α and angiogenic factors were upregulated by the presence of glucose. Ectopically implanted tissue constructs supplemented with glucose exhibited four‐ to fivefold higher viability and were more vascularized compared to those without glucose at day 14. These findings provided the first direct in vitro and in vivo demonstration of the proangiogenic and prosurvival functions of glucose in hMSC upon transplantation and identified glucose as an essential component of the ideal scaffold for transplanting stem cells. STEM CELLS2013;31:526–535


Stem Cells | 2017

Quiescence Preconditioned Human Multipotent Stromal Cells Adopt a Metabolic Profile Favorable for Enhanced Survival under Ischemia

Adrien Moya; Nathanael Larochette; Joseph Paquet; Mickael Deschepper; Morad Bensidhoum; Valentina Izzo; Guido Kroemer; Hervé Petite; Delphine Logeart-Avramoglou

A major impediment to the development of therapies with mesenchymal stem cells/multipotent stromal cells (MSC) is the poor survival and engraftment of MSCs at the site of injury. We hypothesized that lowering the energetic demand of MSCs by driving them into a quiescent state would enhance their survival under ischemic conditions. Human MSCs (hMSCs) were induced into quiescence by serum deprivation (SD) for 48 hours. Such preconditioned cells (SD‐hMSCs) exhibited reduced nucleotide and protein syntheses compared to unpreconditioned hMSCs. SD‐hMSCs sustained their viability and their ATP levels upon exposure to severe, continuous, near‐anoxia (0.1% O2) and total glucose depletion for up to 14 consecutive days in vitro, as they maintained their hMSC multipotential capabilities upon reperfusion. Most importantly, SD‐hMSCs showed enhanced viability in vivo for the first week postimplantation in mice. Quiescence preconditioning modified the energy‐metabolic profile of hMSCs: it suppressed energy‐sensing mTOR signaling, stimulated autophagy, promoted a shift in bioenergetic metabolism from oxidative phosphorylation to glycolysis and upregulated the expression of gluconeogenic enzymes, such as PEPCK. Since the presence of pyruvate in cell culture media was critical for SD‐hMSC survival under ischemic conditions, we speculate that these cells may utilize some steps of gluconeogenesis to overcome metabolic stress. These findings support that SD preconditioning causes a protective metabolic adaptation that might be taken advantage of to improve hMSC survival in ischemic environments. Stem Cells 2017;35:181–196


Tissue Engineering Part A | 2016

Comparison of Survival and Osteogenic Ability of Human Mesenchymal Stem Cells in Orthotopic and Ectopic Sites in Mice.

Mathieu Manassero; Joseph Paquet; Mickael Deschepper; Viateau; Retortillo J; Morad Bensidhoum; Delphine Logeart-Avramoglou; Hervé Petite

Tissue constructs containing mesenchymal stem cells (MSCs) are appealing strategies for repairing large segmental bone defects, but they do not allow consistent bone healing and early cell death was identified as a cause of failure. However, little is known about cell survival in the clinical microenvironment encountered during bone healing process. Osteoconductive coral scaffold with or without luciferase-labeled human MSCs were implanted either in a critical segmental femoral bone defect stabilized by plate or subcutaneously in 44 mice. Cell survival was evaluated by serial bioluminescence imaging (BLI) and osteogenic capabilities by histology and microcomputed tomography. Comparisons between groups were performed with two-way analysis of variance test. Twenty mice were sacrificed 2 weeks after surgery for short-term evaluation and 24 mice at 10 weeks for long-term evaluation. BLI provided evidence of fast and continuous cell death: 85% decrease of the BLI signal over the first 2 weeks in both locations; in fact, less than 2% of the initial cell number was present in all constructs analyzed 4 weeks postimplantation and less than 1% of the initial cell number by 8 weeks postimplantation. By 2 weeks postimplantation, the amount of newly formed bone was self-limited and was similar to ectopic and orthotopic groups. By 10 weeks postimplantation, bone formation was significantly enhanced in the presence of MSCs in orthotopic site and the amount of newly formed bone in cell-containing constructs implanted in orthotopic locations was significantly higher than that observed in the ectopic group. Our results indicated that hMSCs promote bone formation despite early and massive cell death when loaded on coral scaffolds. Interestingly, bone formation was higher in orthotopic than ectopic site despite the same survival pattern. Ectopic implantation of cell-containing constructs is suitable to evaluate cell survival, but assessment of bone formation ability requires orthotopic implantation.


Bio-medical Materials and Engineering | 2012

Impaired osteoblastogenesis potential of progenitor cells in skeletal unloading is associated with alterations in angiogenic and energy metabolism profile.

Katleen Vandamme; Xavier Holy; Morad Bensidhoum; Mickael Deschepper; Delphine Logeart-Avramoglou; Ignace Naert; Joke Duyck; Hervé Petite

Skeletal unloading provokes bone loss. These bone alterations have been shown to be associated with impairment of osteoblastic activity. In the present study, we evaluated the effect of skeletal unloading on bone marrow progenitor cells, for exploration of the underlying mechanism. Wistar rats were randomized to be either hindlimb unloaded for 9 days or to act as controls. Micro-CT was used to detect tibial trabecular architecture changes in response to skeletal unloading. Microgravity conditions for 9 days resulted in a decreased number and an increased spacing of the bone trabeculae in the proximal tibia. The proliferative capacity of the femoral bone marrow samples was assessed (fibroblast-colony-forming assay). By using qPCR, the expression of selected markers of vascularization (Vegfa; Hif1a; Angpt1), energy metabolism (Prkaa2; Mtor), bone formation (Runx2; Alp; Bglap; Bmp2; Bmp4; Bmp7) and bone resorption (Acp5; Tnfsf11; Tnfrsf11b) in these bone marrow suspensions was measured. We demonstrated a striking decrease in the number of fibroblastic progenitors in response to hindlimb unloading. This deficit in proliferation was shown to be accompanied by altered hindlimb perfusion and cellular energy homeostasis. Ex vivo culture assays of the bone marrow-derived progenitor cells screened for osteogenic (Runx2; Alp; Bglap) and adipogenic (Pparg; Fabp4) differentiation alterations in response to microgravity. Induced progenitor cells from unloaded rats showed a delay in osteogenic differentiation and impaired adipogenic differentiation compared to control. The data of this multi-level approach demonstrate that skeletal unloading significantly affects the bone tissue and its metabolism at the progenitor stage. The molecular expressions of the bone marrow population support a role of cellular metabolic stresses in skeletal alterations induced by inactivity.


Stem Cells | 2018

Human Mesenchymal Stem Cell Failure to Adapt to Glucose Shortage and Rapidly Use Intracellular Energy Reserves Through Glycolysis Explains Poor Cell Survival After Implantation

Adrien Moya; Joseph Paquet; Mickael Deschepper; Nathanael Larochette; Karim Oudina; Cyprien Denoeud; Morad Bensidhoum; Delphine Logeart-Avramoglou; Hervé Petite

Mesenchymal stem cells (MSCs) hold considerable promise in tissue engineering (TE). However, their poor survival when exogenously administered limits their therapeutic potential. Previous studies from our group demonstrated that lack of glucose (glc) (but not of oxygen) is fatal to human MSCs because it serves as a pro‐survival and pro‐angiogenic molecule for human MSCs (hMSCs) upon transplantation. However, which energy‐providing pathways MSCs use to metabolize glc upon transplantation? Are there alternative energetic nutrients to replace glc? And most importantly, do hMSCs possess significant intracellular glc reserves for ensuring their survival upon transplantation? These remain open questions at the forefront of TE based‐therapies. In this study, we established for the first time that the in vivo environment experienced by hMSCs is best reflected by near‐anoxia (0.1% O2) rather than hypoxia (1%–5% O2) in vitro. Under these near‐anoxia conditions, hMSCs rely almost exclusively on glc through anerobic glycolysis for ATP production and are unable to use either exogenous glutamine, serine, or pyruvate as energy substrates. Most importantly, hMSCs are unable to adapt their metabolism to the lack of exogenous glc, possess a very limited internal stock of glc and virtually no ATP reserves. This lack of downregulation of energy turnover as a function of exogenous glc level results in a rapid depletion of hMSC energy reserves that explains their poor survival rate. These new insights prompt for the development of glc‐releasing scaffolds to overcome this roadblock plaguing the field of TE based‐therapies. Stem Cells 2018;36:363–376


Tissue Engineering Part A | 2013

Bone Regeneration in Sheep Using Acropora Coral, a Natural Resorbable Scaffold, and Autologous Mesenchymal Stem Cells

Mathieu Manassero; Véronique Viateau; Mickael Deschepper; Karim Oudina; Delphine Logeart-Avramoglou; Hervé Petite; Morad Bensidhoum


Tissue Engineering Part C-methods | 2011

A perfusion bioreactor for engineering bone constructs: an in vitro and in vivo study.

Bertrand David; Dominique Bonnefont-Rousselot; Karim Oudina; Marie-Christelle Degat; Mickael Deschepper; Véronique Viateau; Morad Bensidhoum; Christian Oddou; Hervé Petite


Tissue Engineering Part C-methods | 2013

A Novel Murine Femoral Segmental Critical-Sized Defect Model Stabilized by Plate Osteosynthesis for Bone Tissue Engineering Purposes

Mathieu Manassero; Véronique Viateau; Romano Matthys; Mickael Deschepper; Rosario Vallefuoco; Morad Bensidhoum; Hervé Petite


Ref : TIP597WEB - "Technologies biomédicales" | 2015

Des biomatériaux à l'ingénierie tissulaire : perspectives en réparation osseuse

Mickael Deschepper; Joseph Paquet; Adrien Moya; Mathieu Manassero; Morad Bensidhoum; Hervé Petite

Collaboration


Dive into the Mickael Deschepper's collaboration.

Top Co-Authors

Avatar

Hervé Petite

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Delphine Logeart-Avramoglou

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Joseph Paquet

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Mathieu Manassero

École nationale vétérinaire d'Alfort

View shared research outputs
Top Co-Authors

Avatar

Karim Oudina

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Adrien Moya

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Véronique Viateau

École nationale vétérinaire d'Alfort

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Laurent‐Emmanuel Monfoulet

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Marie-Christelle Degat

Centre national de la recherche scientifique

View shared research outputs
Researchain Logo
Decentralizing Knowledge