40 research outputs found

    alpha-Tocopherol Acetate Attenuates Mitochondrial Oxygen Consumption and Maintains Primitive Cells within Mesenchymal Stromal Cell Population

    Get PDF
    We present here the data showing, in standard cultures exposed to atmospheric O-2 concentration, that alpha-tocopherol acetate (alpha-TOA) has a positive impact on primitive cells inside mesenchymal stromal cell (MstroC) population, by maintaining their proliferative capacity. alpha-TOA decreases the O-2 consumption rate of MStroC probably by impacting respiratory chain complex II activity. This action, however, is not associated with a compensatory increase in glycolysis activity, in spite of the fact that the degradation of HIF-1 alpha was decreased in presence of alpha-TOA. This is in line with a moderate enhancement of mtROS upon alpha-TOA treatment. However, the absence of glycolysis stimulation implies the inactivity of HIF-1 alpha which might - if it were active - be related to the maintenance of stemness. It should be stressed that alpha-TOA might act directly on the gene expression as well as the mtROS themselves, which remains to be elucidated.This is the peer reviewed version of the paper: Loncarić, D., Rodriguez, L., Debeissat, C., Touya, N., Labat, V., Villacreces, A., Bouzier-Sore, A.-K., Pasquet, J.-M., de la Grange, P. B., Vlaski-Lafarge, M., Pavlović, S., & Ivanović, Z. (2021). Alpha-Tocopherol Acetate Attenuates Mitochondrial Oxygen Consumption and Maintains Primitive Cells within Mesenchymal Stromal Cell Population. Stem Cell Reviews and Reports, 17(4), 1390–1405.[ https://doi.org/10.1007/s12015-020-10111-9]Related to published version: [https://imagine.imgge.bg.ac.rs/handle/123456789/1491

    Régulation de l'hématopoïèse par les basses concentrations d'oxygène (rôles de l'antigène CD34 et du facteur de croissance VEGF165)

    No full text
    L'hématopoïèse, processus de production des cellules sanguines à partir des cellules souches, est notamment régulée par les concentrations d'O2 médullaires comprises entre 0 à 5 % (hypoxie). Nous avons établi des liens entre des facteurs intrinsèques à la cellule impliqués dans le processus de différenciation (antigène CD34) et leur sensibilité à l'hypoxie, et étudié les effets sur l'hématopoïèse de facteurs inductibles par l'hypoxie (Vascular Endothelial Growth Factor, VEGF). La culture de cellules hématopoiétiques CD34+ à 1 % d'O2 augmente ou stabilise l'expression du gène cd34 qui diminue à 20 % d'O2. Le maintien prolongé de cette expression associé à une expression membranaire durable de la protéine sont corrélés avec le statut immature des cellules. D'autre part, le VEGF165 permet la survie de cellules souches murines en culture liquide à 1 % d'O2. Ainsi l'hypoxie freine la différenciation des cellules souches par l'expression du gène cd34 et favorise leur survie par le VEGF165.Hematopoiesis, the process of mature blood production from stem cells, is in part regulated by bone marrow oxygen concentrations, which vary from 0 to 5 % (hypoxia). We studied in this work the relationships between cell intrinsic factors involved in the maturation process (CD34 antigen) and their sensitivity to hypoxia, and the effects of molecules inducible by hypoxia (Vascular Endothelial Growth Factor, VEGF) on hematopoiesis. We showed that cultures of CD34+ cells at 1 % O2 induce or stabilize the cd34 gene expression that decreases at 20 % O2. The prolonged maintenance of this expression associated with the long-lasting membrane expression of the protein were correlated with the primitiveness of cells. We also showed that VEGF165 led to the survival of murine stem cells cultured at 1 % O2. This work suggests that hypoxia slows down the differentiation of stem cells by inducing cd34 gene expression, and favours their survival through VEGF165.BORDEAUX2-BU Santé (330632101) / SudocPARIS-BIUP (751062107) / SudocSudocFranceF

    Immature hematopoietic stem cells undergo maturation in the fetal liver.

    Get PDF
    International audienceHematopoietic stem cells (HSCs), which are defined by their capacity to reconstitute adult conventional mice, are first found in the dorsal aorta after 10.5 days post coitus (dpc) and in the fetal liver at 11 dpc. However, lympho-myeloid hematopoietic progenitors are detected in the dorsal aorta from 9 dpc, raising the issue of their role in establishing adult hematopoiesis. Here, we show that these progenitors are endowed with long-term reconstitution capacity, but only engraft natural killer (NK)-deficient Rag2γc(-/-) mice. This novel population, called here immature HSCs, evolves in culture with thrombopoietin and stromal cells, into HSCs, defined by acquisition of CD45 and MHC-1 expression and by the capacity to reconstitute NK-competent mice. This evolution occurs during ontogeny, as early colonization of fetal liver by immature HSCs precedes that of HSCs. Moreover, organ culture experiments show that immature HSCs acquire, in this environment, the features of HSCs

    Targeting the aryl hydrocarbon receptor nuclear translocator complex with DMOG and Stemregenin 1 improves primitive hematopoietic stem cell expansion

    Get PDF
    Culture conditions used for the expansion of hematopoietic stemand progenitor cells (HSCs andHPCs, collectively HSPCs) should ideally favor the self renewal of long-termHSCs. At 20% O2, the synthesis of HIF-1α is balanced by its hydroxylation and proteasomal degradation. This favors HSPC differentiation, but can be prevented by culturing CD34+cord blood cells in the presence of dimethyloxaloylglycine (DMOG). This differentiation may also be reduced by culturing the cells in the presence of Stemregenin 1, an antagonist of the aryl hydrocarbon receptor (AhR). The objective of this studywas to investigate howhypoxia,DMOG and Stemregenin 1might affect the expansion ofHSPCswith the aim of identifying optimal conditions for expansion in culture. It was found thatDMOG decreased proliferation but was effective in preserving the number of cells in the primitive hematopoietic subpopulations in vitro. The effect of DMOG was similar to hypoxia, although differenceswere observedwith regard to the side population and CD34+ sub-populations. Stemregenin 1 on the other hand increased the size of the primitive as well as the other HSC sub-populations. The use of Stemregenin 1 with DMOG increased the proportion of primitiveHSCs to 3.54% compared to 2.61% for Stemregenin 1 alone. In vivo engraftment studies confirmed these findings and showed that fewer cells (3710) are required for long-term engraftmentwhen HSCs are grown in Stemregenin 1 together with hypoxia than in Stemregenin 1 under conditions of normoxia (13430).The Medical Research Council of South Africa in terms of the MRC's Flagships Awards Project SAMRC-RFA-UFSP-01-2013/STEM CELLS, the SAMRC Extramural Unit for Stem Cell Research and Therapy, the Institute for Cellular and Molecular Medicine of the University of Pretoria, Etablissement Français du Sang, the University of Bordeaux fellowship, the German Academic Exchange Service and the National Research Foundation of South Africa (DAAD-NRF).http://www.elsevier.com/locate/scrBiochemistryImmunolog

    Alpha Lipoic-Acid Potentiates Ex Vivo Expansion of Human Steady-State Peripheral Blood Hematopoietic Primitive Cells

    No full text
    Steady state peripheral blood (SSPB) contains hematopoietic stem and progenitor cells (HSPCs) presenting characteristics of real hematopoietic stem cells, and thus represents an interesting alternative cell supply for hematopoietic cell transplantation. Development of ex vivo expansion strategies could overcome the low HSPC numbers usually rescued from SSPB. We investigated the effect of alpha lipoic acid (ALA) on ex vivo culture of SSPB CD34 positive (CD34pos) cells on primitive cell expansion, cell cycle, and oxidative metabolism as estimated by determining the ROS and GSH content. ALA increased the ex vivo expansion of total CD34pos cells and of phenotypically defined CD34pos HSPCs subpopulations that retained in vivo repopulating capacity, concomitantly to a decreased expansion of differentiating cells. ALA did not modify cell cycle progression nor the proliferation of ex vivo expanded CD34pos cells, and coherently did not affect the ROS level. On the contrary, ALA decreased the proliferation and disturbed cell cycle progression of cells reaching a differentiated status, a phenomenon that seems to be associated with a drop in ROS level. Nonetheless, ALA affected the redox status of hematopoietic primitive cells, as it reproducibly increased GSH content. In conclusion, ALA represents an interesting molecule for the improvement of ex vivo expansion strategies and further clinical application in hematopoietic cell transplantation (HCT)

    Repopulating Hematopoietic Stem Cells From Steady-State Blood Before And After Ex Vivo Culture Are Enriched In CD34+CD133+CXCR4low Fraction

    No full text
    The feasibility of ex vivo expansion allows us to consider the steady-state peripheral blood as an alternative source of hematopoietic stem progenitor cells for transplantation when growth factor-induced cell mobilization is contraindicated or inapplicable. Ex vivo expansion dramatically enhances the in vivo reconstituting cell population from steady-state blood. In order to investigate phenotype and the expression of homing molecules, the expression of CD34, CD133, CD90, CD45RA, CD26 and CD9 was determined on sorted CD34+ cells according to CXCR4 (“neg”, “low” “bright”) and CD133 expression before and after ex vivo expansion. Hematopoietic stem cell activity was determined in vivo on the basis of hematopoietic repopulation of primary and secondary recipients - NSG immuno-deficient mice. In vivo reconstituting cells in the steady-state blood CD34+ cell fraction before expansion belong to the CD133+ population and are CXCR4low or, to a lesser extent, CXCR4neg, while after ex vivo expansion they are contained only in the CD133+CXCR4low cells. The failure of the CXCR4bright population to engraft is probably due to the exclusive expression of CD26 by these cells. The limiting-dilution analysis showed that both repopulating cell number and individual proliferative capacity were enhanced by ex vivo expansion. Thus, steady-state peripheral blood cells exhibit a different phenotype compared to mobilized and cord blood cells, as well as to those issued from the bone marrow. These data represent the first phenotypic characterization of steady-state blood cells exhibiting short- and long-term hematopoietic reconstituting potential, which can be expanded ex vivo, a sine qua non for their subsequent use for transplantation.publishedVersionCopyright © 2018 Ferrata Storti Foundation Material published in Haematologica is covered by copyright. All rights are reserved to the Ferrata Storti Foundation. Use of published material is allowed under the following terms and conditions: https://creativecommons.org/licenses/by-nc/4.0/legalcod

    Busulfan administration flexibility increases the applicability of scid repopulating cell assay in NSG mouse model.

    Get PDF
    BACKGROUND: Xenotransplantation models allowing the identification and quantification of human Hematopoietic stem cells (HSC) in immunodeficient mice remain the only way to appropriately address human HSC function despite the recent progress in phenotypic characterization. However, these in vivo experiments are technically demanding, time consuming and expensive. Indeed, HSCs engraftment in mouse requires pre-conditioning of animals either by irradiation or cytotoxic drugs to allow homing of injected cells in specific stem cell niches and their subsequent expansion and differentiation in bone marrow. Recently, the development of busulfan pre-conditioning of animals improved the flexibility of experimentation in comparison with irradiation. DESIGN AND METHODS: In order to further facilitate the organization of these complex experiments we investigated the effect of extending the period between mice pre-conditioning and cell injection on the engraftment efficiency. In the meantime, we also explored the role of busulfan doses, mouse gender and intravenous injection route (caudal or retro orbital) on engraftment efficiency. RESULTS AND CONCLUSION: We showed that a period of up to 7 days did not modify engraftment efficiency of human HSCs in NSG model. Moreover, retro orbital cell injection to female mice pre-conditioned with 2x25 mg/kg of busulfan seems to be the best adapted schema to detect the human HSC in xenotransplantation experiments
    corecore