Network


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

Hotspot


Dive into the research topics where Olivier Giet is active.

Publication


Featured researches published by Olivier Giet.


Transfusion | 2013

Infusion of clinical-grade enriched regulatory T cells delays experimental xenogeneic graft-versus-host disease.

Muriel Hannon; Chantal Lechanteur; Sophie Lucas; Joan Somja; Laurence Seidel; Ludovic Belle; Etienne Baudoux; Olivier Giet; André-Marie Chantillon; Philippe Delvenne; Pierre Drion; Yves Beguin; Stéphanie Humblet-Baron; Frédéric Baron

We investigated the ability of clinical‐grade enriched human regulatory T cells (Treg) to attenuate experimental xenogeneic graft‐versus‐host disease (GVHD) induced by peripheral blood mononuclear cells (PBMNCs; autologous to Treg) infusion in NSG mice, as well as verified their inability to induce xenogeneic GVHD when infused alone.


Experimental Hematology | 2001

Cell cycle activation of hematopoietic progenitor cells increases very late antigen-5–mediated adhesion to fibronectin

Olivier Giet; Sandra Huygen; Yves Beguin; André Gothot

Recent studies suggested that trafficking of hematopoietic progenitor cells is related to cell cycle status. We studied whether adhesion of progenitor cells to extracellular matrix proteins was modulated by cell cycle transit. Mobilized peripheral blood CD34+ cells were stimulated ex vivo for 48 hours with stem cell factor, flt-3 ligand, and thrombopoietin and fractionated by adhesion to fibronectin or vascular cell adhesion molecule-1 (VCAM-1). Adherent and nonadherent cells were assayed for cell cycle status, long-term culture-initiating cell frequency, and integrin function. Binding to fibronectin, but not to VCAM-1, displayed a cell cycle selectivity as the adherent fraction to fibronectin was enriched in cycling CD34+ cells and in cycling long-term culture-initiating cells compared to the nonadherent fraction. Combined cell cycle and phenotypic analysis showed that the expression of VLA-5 was upregulated during S/G2+M but that of VLA-4 remained constant. The selective binding of cycling CD34+ cells to fibronectin was reverted by anti-VLA-5 but not by anti-VLA-4 blocking antibodies. Also, cycling CD34+ cells preferentially adhered to the VLA-5 binding domain but not to the VLA-4 binding domain of fibronectin. Adhesion of cycling CD34+ cells to fibronectin was a reversible process modulated by cell cycle progression, because adherent cells could exit the cell cycle and return to a nonadhesive state within an additional 48-hour culture period. The results indicate that the enhanced binding capacity of cycling progenitor cells to fibronectin is mediated by VLA-5.


Transplantation | 2014

Impact of Cotransplantation of Mesenchymal Stem Cells on Lung Function After Unrelated Allogeneic Hematopoietic Stem Cell Transplantation Following Non-Myeloablative Conditioning

Catherine Moermans; Chantal Lechanteur; Etienne Baudoux; Olivier Giet; Monique Henket; Laurence Seidel; Marie Lejeune; Evelyne Willems; Frédéric Baron; Renaud Louis; Yves Beguin

Background In the context of hematopoietic stem cell transplantation (HSCT), mesenchymal stem cells (MSC) have been used to promote engraftment and prevent graft-versus-host disease. However, in animal models, MSC were shown to cause pulmonary alterations after systemic administration. The impact of MSC infusion on lung function has not been studied in humans. The objective of the study was to investigate the impact of MSC co-infusion on lung function and airway inflammation as well as on the incidence of pulmonary infections and cytomegalovirus (CMV) reactivation after HSCT. Methods We have prospectively followed 30 patients who underwent unrelated HSCT with MSC co-infusion after non-myeloablative conditioning (NMA). Each patient underwent detailed lung function testing (FEV1, FVC, FEV1/FVC, RV, TLC, DLCO, and KCO) and measurement of exhaled nitric oxide before HSCT and 3, 6, and 12 months posttransplant. The incidence of pulmonary infections and CMV reactivation were also monitored. This group was compared with another group of 28 patients who underwent the same type of transplantation but without MSC co-infusion. Results Lung function tests did not show important modifications over time and did not differ between the MSC and control groups. There was a higher 1-year incidence of infection, particularly of fungal infections, in patients having received a MSC co-infusion. There was no difference between groups regarding the 1-year incidence of CMV reactivation. Conclusions MSC co-infusion does not induce pulmonary deterioration 1 year after HSCT with NMA conditioning. MSC appear to be safe for the lung, but close monitoring of pulmonary infections remains essential.


Stem Cells | 2006

Despite Inhibition of Hematopoietic Progenitor Cell Growth In Vitro, the Tyrosine Kinase Inhibitor Imatinib Does Not Impair Engraftment of Human CD133+ Cells into NOD/SCIDβ2mNull Mice

Laurence Pirson; Frédéric Baron; Nathalie Meuris; Olivier Giet; Emilie Castermans; Roland Greimers; Ivano Di Stefano; André Gothot; Yves Beguin

There is potential interest for combining allogeneic hematopoietic cell transplantation (HCT), and particularly allogeneic HCT with a nonmyeloablative regimen, to the tyrosine kinase inhibitor imatinib (Glivec; Novartis, Basel, Switzerland, http://www.novartis.com) in order to maximize anti‐leukemic activity against Philadelphia chromosome‐positive leukemias. However, because imatinib inhibits c‐kit, the stem cell factor receptor, it could interfere with bone marrow engraftment. In this study, we examined the impact of imatinib on normal progenitor cell function. Imatinib decreased the colony‐forming capacity of mobilized peripheral blood human CD133+ cells but not that of long‐term culture‐initiating cells. Imatinib also decreased the proliferation of cytokine‐stimulated CD133+ cells but did not induce apoptosis of these cells. Expression of very late antigen (VLA)‐4, VLA‐5, and CXCR4 of CD133+ cells was not modified by imatinib, but imatinib decreased the ability of CD133+ cells to migrate. Finally, imatinib did not decrease engraftment of CD133+ cells into irradiated nonobese diabetic/severe combined immunodeficient/β2mnull mice conditioned with 3 or 1 Gy total body irradiation. In summary, our results suggest that, despite inhibition of hematopoietic progenitor cell growth in vitro, imatinib does not interfere with hematopoietic stem cell engraftment.


Haematologica | 2009

Ex vivo expansion of hematopoietic progenitor cells is associated with downregulation of alpha4 integrin- and CXCR4-mediated engraftment in NOD/SCID beta2-microglobulin-null mice

Jacques Foguenne; Ivano Di Stefano; Olivier Giet; Yves Beguin; André Gothot

This study shows that ex vivo culture of hematopoietic stem cells is associated with downregulation of both α4 and CXCR4-mediated engraftment. Background Several studies indicate that ex vivo cytokine-supported expansion induces defective hematopoietic stem cell engraftment. We investigated the role of α4 integrin, α5 integrin and CXCR4 in engraftment of unmanipulated and cytokine-treated human cord blood CD34+ cells. Design and Methods Uncultured or expanded CD34+ cells were infused in NOD/SCID-β2microglobulin-null mice. The function of α4, and α5 integrins and CXCR4 was assessed by incubating cells with specific neutralizing antibodies, prior to transplant. The activation state of α4 integrin was further tested by adhesion and migration assays. Results Neutralization of either α4 integrin or CXCR4 abolished engraftment of uncultured CD34+ cells at 6 week spost-transplant, while α5 integrin neutralization had no significant effect. However, after short-term ex vivo culture, blocking α4 integrin or CXCR4 did not affect repopulating activity whereas neutralization of α5 integrin inhibited engraftment. Using soluble vascular cell adhesion molecule-1 binding assays, we observed that α4 integrin affinity in fresh CD34+ cells was low and susceptible to stimulation while in cultured CD34+ cells, it was high and insensitive to further activation. In addition, stromal cell-derived factor-1 stimulated migration across vascular cell adhesion molecule-1 in fresh CD34+ cells but not in cultured CD34+ cells. Conclusions Our data show that ex vivo culture of hematopoietic progenitor cells is associated with downregulation of both α4 integrin- and CXCR4-mediated engraftment. Further investigations suggest that this is caused by supraphysiological increase of α4 integrin affinity, which impairs directional migration across vascular cell adhesion molecule-1 in response to stromal cell-derived factor-1. Such changes may underlie the engraftment defect of cytokine-stimulated CD34+ cells.


Journal of Hepatology | 2017

Infusion of mesenchymal stromal cells after deceased liver transplantation: A phase I-II, open-label, clinical study.

Olivier Detry; Morgan Vandermeulen; Marie-Hélène Delbouille; Joan Somja; Noëlla Bletard; Alexandra Briquet; Chantal Lechanteur; Olivier Giet; Etienne Baudoux; Muriel Hannon; Frédéric Baron; Yves Beguin

BACKGROUND & AIMS Mesenchymal stromal cell (MSC) infusion could be a means to establish tolerance in solid organ recipients. The aim of this prospective, controlled, phase I study was to evaluate the feasibility, safety and tolerability of a single infusion of MSCs in liver transplant recipients. METHODS Ten liver transplant recipients under standard immunosuppression received 1.5-3×106/kg third-party unrelated MSCs on postoperative day 3±2, and were prospectively compared to a control group of ten liver transplant recipients. As primary endpoints, MSC infusion toxicity was evaluated, and infectious and cancerous complications were prospectively recorded until month 12 in both groups. As secondary endpoints, rejection rate, month-6 graft biopsies, and peripheral blood lymphocyte phenotyping were compared. Progressive immunosuppression weaning was attempted from month 6 to 12 in MSC recipients. RESULTS No variation in vital parameters or cytokine release syndrome could be detected during and after MSC infusion. No patient developed impairment of organ functions (including liver graft function) following MSC infusion. No increased rate of opportunistic infection or de novo cancer was detected. As secondary endpoints, there was no difference in overall rates of rejection or graft survival. Month-6 biopsies did not demonstrate a difference between groups in the evaluation of rejection according to the Banff criteria, in the fibrosis score or in immunohistochemistry (including Tregs). No difference in peripheral blood lymphocyte typing could be detected. The immunosuppression weaning in MSC recipients was not successful. CONCLUSIONS No side effect of MSC infusion at day 3 after liver transplant could be detected, but this infusion did not promote tolerance. This study opens the way for further MSC or Treg-based trials in liver transplant recipients. LAY SUMMARY Therapy with mesenchymal stromal cells (MSCs) has been proposed as a means to improve results of solid organ transplantation. One of the potential MSC role could be to induce tolerance after liver transplantation, i.e. allowing the cessation of several medications with severe side effects. This study is the first-in-man use of MSC therapy in ten liver transplant recipients. This study did not show toxicity after a single MSC infusion but it was not sufficient to allow withdrawal of immunosuppression. CLINICAL TRIAL REGISTRATION NUMBER Eudract: # 2011-001822-81, ClinicalTrials.gov: # NCT 01429038.


Pathologie Biologie | 2014

Quality controls on cord blood unit contiguous segments: recommendation of the SFGM-TC.

J. De Vos; B. Birebent; Catherine Faucher; Olivier Giet; Y. Hicheri; C. Lemarie; Etienne Baudoux; Florence Boulanger; F. Garnier; J. Larghero; Fabienne Pouthier; Jean-Baptiste Thibert; Ibrahim Yakoub-Agha

In the attempt to harmonize clinical practices between different French transplantation centers, the French Society of Bone Marrow Transplantation and Cell Therapies (SFGM-TC) set up its fourth annual series of workshops which brought together practitioners from all of its member centers. These workshops took place in September 2013 in Lille. Literature and intra-laboratories studies suggest that attached segment is representative of cord blood unit (CBU). Nevertheless, some discrepancies have been observed when analyzing large data registries. To address these issues, we have listed recommendations to increase the standardization of segment processing and quality control (QC), information on units of measurement and specifications and action to be taken in case of out of specifications QC results on segment.


Current Hematologic Malignancy Reports | 2014

Thinking Out of the Box—New Approaches to Controlling GVHD

Frédéric Baron; Stéphanie Humblet-Baron; Grégory Ehx; Sophie Servais; Muriel Hannon; Ludovic Belle; Chantal Lechanteur; Alexandra Briquet; Olivier Giet; Etienne Baudoux; Evelyne Willems; Yves Beguin

Graft-versus-host disease (GVHD) remains a major limitation of allogeneic hematopoietic cell transplantation (allo-HCT). Despite major advances in the understanding of GVHD pathogenesis, standard GVHD prophylaxis regimens continue to be based on the combination of a calcineurin inhibitor with an antimetabolite, while first line treatments still rely on high-dose corticosteroids. Further, no second line treatment has emerged thus far in acute or chronic GVHD patients who failed to respond with corticosteroid treatment. After briefly reviewing current standards of GVHD prevention and treatment, this article will discuss recent approaches that might change GVHD prophylaxis/treatment for decades to come, with a special focus on recently developed immunoregulatory strategies based on infusion of mesenchymal stromal or regulatory T-cells, or injection of low-dose interleukin-2.


Experimental Hematology | 2000

Upregulation of VLA-5 during cell cycle transit prevents effective migration of CD34+ cells through fibronectin-coated filters

Olivier Giet; Sandra Huygen; Yves Beguin; André Gothot

Abstract Several reports have suggested that engraftment of stem/progenitor cells is decreased during cell cycle transit. This prompted us to study cell cycle-associated changes in adhesion and migration after a 2-day ex vivo culture in SCF, FL and TPO. We previously demonstrated that VLA-5 expression of CD34+ cells was enhanced during cell cycle transit and mediated increased adhesion to fibronectin (Fn). Here, we examined whether the increase in VLA-5 mediated adhesion to Fn had any impact on transmigration of cord blood CD34+ cells. Migration was assayed on 5-μm pore Transwells during 3 hours. Migration towards control medium through BSA-coated filters was observed in 5% of the cells. An additional 15% of input cells migrated when the filter was coated with Fn. Migration across Fn-coated filters towards conditioned medium (CM) from the stromal-derived factor (SDF)-1 producing cell line MS-5 rose to 42%. Migration was dependent on the presence of CXCR4, as 48% of migrating (Mg) cells were CXCR4+ versus 20% of non migrating (NMg) cells (P


Blood | 2002

Adhesion of synchronized human hematopoietic progenitor cells to fibronectin and vascular cell adhesion molecule-1 fluctuates reversibly during cell cycle transit in ex vivo culture.

Sandra Huygen; Olivier Giet; Vincent Artisien; Ivano Di Stefano; Yves Beguin; André Gothot

Collaboration


Dive into the Olivier Giet's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge