Network


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

Hotspot


Dive into the research topics where Christelle Demarquay is active.

Publication


Featured researches published by Christelle Demarquay.


Journal of Gene Medicine | 2003

Mesenchymal stem cells home to injured tissues when co-infused with hematopoietic cells to treat a radiation-induced multi-organ failure syndrome

Alain Chapel; Jean Marc Bertho; Morad Bensidhoum; Loic Fouillard; Randell G. Young; Johanna Frick; Christelle Demarquay; Frédérique Cuvelier; Emilie Mathieu; F. Trompier; Nicolas Dudoignon; Claire Germain; Christelle Mazurier; Jocelyne Aigueperse; Jade Borneman; Norbert Claude Gorin; Patrick Gourmelon; Dominique Thierry

Recent studies have suggested that ex vivo expansion of autologous hematopoietic cells could be a therapy of choice for the treatment of bone marrow failure. We investigated the potential of a combined infusion of autologous ex vivo expanded hematopoietic cells with mesenchymal (MSCs) for the treatment of multi‐organ failure syndrome following irradiation in a non‐human primate model.


PLOS ONE | 2013

Mesenchymal stem cell therapy stimulates endogenous host progenitor cells to improve colonic epithelial regeneration.

Alexandra Sémont; Christelle Demarquay; Raphaëlle Bessout; Christelle Durand; Marc Benderitter; Noëlle Mathieu

Patients who undergo pelvic radiotherapy may develop severe and chronic complications resulting from gastrointestinal alterations. The lack of curative treatment highlights the importance of novel and effective therapeutic strategies. We thus tested the therapeutic benefit of mesenchymal stem cells (MSC) treatment and proposed molecular mechanisms of action. MSC efficacy was tested in an experimental model of radiation-induced severe colonic ulceration histologically similar to that observed in patients. In this model, MSC from bone marrow were administered intravenously, immediately or three weeks (established lesions) after irradiation. MSC therapy reduces radiation-induced colonic ulceration and increases animal survival. MSC treatment induces therapeutic efficacy whatever the time of cell infusion. Infused-MSC engraft in the colon but also increase endogenous MSC mobilization in blood that have lasting benefits over time. In vitro analysis demonstrates that the MSC effect is mediated by paracrine mechanisms through the non-canonical WNT (Wingless integration site) pathway. In irradiated rat colons, MSC treatment increases the expression of the non-canonical WNT4 ligand by epithelial cells. The epithelial regenerative process is improved after MSC injection by stimulation of colonic epithelial cells positive for SOX9 (SRY-box containing gene 9) progenitor/stem cell markers. This study demonstrates that MSC treatment induces stimulation of endogenous host progenitor cells to improve the regenerative process and constitutes an initial approach to arguing in favor of the use of MSC to limit/reduce colorectal damage induced by radiation.


Radiation Research | 2005

Radiation-Induced Increase in Plasma Flt3 Ligand Concentration in Mice: Evidence for the Implication of Several Cell Types

Marie Prat; Christelle Demarquay; Johanna Frick; Dominique Thierry; Norbert-Claude Gorin; Jean Marc Bertho

Abstract Prat, M., Demarquay, C., Frick, J., Thierry, D., Gorin, N. C. and Bertho, J. M. Radiation-Induced Increase in Plasma Flt3 Ligand Concentration in Mice: Evidence for the Implication of Several Cell Types. Radiat. Res. 163, 408–417 (2005). Circulating T lymphocytes were proposed as the main producer of Flt3 ligand. However, during aplasia, there is a drastic reduction in the number of T lymphocytes, while plasma Flt3 ligand concentration is increased. This contradiction prompted us to compare variations in plasma Flt3 ligand during radiation-induced aplasia in BALB/c mice and in T-lymphocyte-deficient NOD-SCID mice to delineate the role of T lymphocytes in the increase in Flt3 ligand concentration. The results showed that plasma Flt3 ligand concentration was increased similarly in the two strains of mice, and that Flt3 ligand concentration was negatively correlated to the number of residual hematopoietic progenitors. Moreover, the Flt3 ligand mRNA expression and Flt3 ligand protein concentration were similar in the two strains of mice in all organs tested, i.e. thymus, spleen, bone marrow, liver, brain and blood cells. These results confirm that Flt3 ligand concentration in the blood is a reflection of bone marrow function and that T lymphocytes are not the main regulator of Flt3 ligand variations during aplasia.


Radiation Research | 2006

Use of Flt3 Ligand to Evaluate Residual Hematopoiesis after Heterogeneous Irradiation in Mice

Marie Prat; Christelle Demarquay; Johanna Frick; Nicolas Dudoignon; Dominique Thierry; Jean Marc Bertho

Abstract Prat, M., Demarquay, C., Frick, J., Dudoignon, N., Thierry, D. and Bertho, J. M. Use of Flt3 Ligand to Evaluate Residual Hematopoiesis after Heterogeneous Irradiation in Mice. Radiat. Res. 166, 504–511 (2006). We evaluated the possibility of using plasma Flt3 ligand (FL) concentration as a biological indicator of bone marrow function after heterogeneous irradiation. Mice were irradiated with 4, 7.5 or 11 Gy with 25, 50, 75 or 100% of the bone marrow in the field of irradiation. This model of irradiation resulted in graded and controlled damage to the bone marrow. Mice exhibited a pancytopenia correlated with both the radiation dose and the percentage of bone marrow irradiated. The FL concentration in the blood increased with the severity of bone marrow aplasia. Nonlinear regression analysis showed that the FL concentration was strongly correlated with the total number of residual colony-forming cells 3 days after irradiation, allowing a precise estimate of residual hematopoiesis. Moreover, the FL concentration on day 3 postirradiation was correlated with the duration and severity of subsequent pancytopenia, suggesting that variations in FL concentrations might be used as a predictive indicator of bone marrow aplasia, especially by the use of linear regression equations describing these correlations. Our results provide a rationale for the use of FL concentration as a biological indicator of residual hematopoiesis after heterogeneous irradiation.


Radiation Research | 2005

Application of Autologous Hematopoietic Cell Therapy to a Nonhuman Primate Model of Heterogeneous High-Dose Irradiation

Jean-Marc Bertho; Marie Prat; Johanna Frick; Christelle Demarquay; Marie-Hélène Gaugler; Nicolas Dudoignon; I. Clairand; Alain Chapel; Norbert-Claude Gorin; Dominique Thierry; Patrick Gourmelon

Abstract Bertho, J-M., Prat, M., Frick, J., Demarquay, C., Gaugler, M-H., Dudoignon, N., Clairand, I., Chapel, A., Gorin, N-C., Thierry, D. and Gourmelon, P. Application of Autologous Hematopoietic Cell Therapy to a Nonhuman Primate Model of Heterogeneous High-Dose Irradiation. Radiat. Res. 163, 557– 570 (2005). We developed a model of heterogeneous irradiation in a nonhuman primate to test the feasibility of autologous hematopoietic cell therapy for the treatment of radiation accident victims. Animals were irradiated either with 8 Gy to the body with the right arm shielded to obtain 3.4 Gy irradiation or with 10 Gy total body and 4.4 Gy to the arm. Bone marrow mononuclear cells were harvested either before irradiation or after irradiation from an underexposed area of the arm and were expanded in previously defined culture conditions. We showed that hematopoietic cells harvested after irradiation were able to expand and to engraft when reinjected 7 days after irradiation. Recovery was observed in all 8-Gy-irradiated animals, and evidence for a partial recovery was observed in 10-Gy-irradiated animals. However, in 10-Gy-irradiated animals, digestive disease was observed from day 16 and resulted in the death of two animals. Immunohistological examinations showed damage to the intestine, lungs, liver and kidneys and suggested radiation damage to endothelial cells. Overall, our results provide evidence that such an in vivo model of heterogeneous irradiation may be representative of accidental radiation exposures and may help to define the efficacy of therapeutic interventions such as autologous cell therapy in radiation accident victims.


Stem Cells and Development | 2008

Characterization and Histological Localization of Multipotent Mesenchymal Stromal Cells in the Human Postnatal Thymus

Moubarak Mouiseddine; Noëlle Mathieu; Johanna Stefani; Christelle Demarquay; Jean-Marc Bertho

The aim of this work was to characterize multipotent mesenchymal stromal cells (MSCs) in the postnatal human thymus and to localize these MSCs in the organ. Adherent cells isolated from thymus samples were characterized by cell-surface antigen expression. This showed that adherent cells have a MSC profile as assessed by the expression of CD73 and CD105 markers and the lack of CD45 expression. These cells are able to differentiate in vitro into adipocytes, osteoblasts, and chondrocytes and to inhibit mixed lymphocyte reaction. This indicates that isolated cells have all of the characteristics of MSC. The fibroblast colony-forming unit (CFU-F) assay was used to determine their frequency in the postnatal thymus. This frequency was 60.9 +/- 14.8 CFU-F per 1 x 10(5) freshly isolated mononuclear cells. Moreover, taking advantage of CD34 and CD105 expression, immunohistological staining allowed us to localize MSC within interlobular trabeculae in close contact with the outer cortex. Polymerase chain reaction experiments indicated that thymic MSC expressed interleukin-7 and stromal cell-derived factor-1 messenger RNA. Overall, these results confirm previous findings of the presence in the adult human thymus of multipotent MSCs with a phenotype similar to adipose-derived adult stem cells. These results also show for the first time a histological localization of MSC in an organ. This suggests a possible role of thymic MSC in intrathymic differentiation.


International Journal of Radiation Biology | 2004

Feasibility and limits of bone marrow mononuclear cell expansion following irradiation

Jean-Marc Bertho; E. Mathieu; A. Lauby; Johanna Frick; Christelle Demarquay; Patrick Gourmelon; N. C. Gorin; Dominique Thierry

Purpose: To define the ability of bone marrow mononuclear cells (BMMNC) to expand after irradiation and to determine the amount of apoptosis in irradiated expanded cells. Materials and methods: Non‐human primate BMMNC were irradiated in vitro at doses ranging from 0 to 4 Gy and were cultured during 1 week in the presence of interleukin 3, interleukin 6, stem cell factor, thrombopoietin and fms‐like tyrosine kinase‐3 ligand. The expansion yield of BMMNC, colony‐forming cells and CD34+ cells were compared with non‐irradiated control cultures. Apoptosis in expanded cells was also defined by annexin V/propidium iodine staining. Results: Irradiation of BMMNC up to 1 Gy did not modify the ability of haematopoietic cells to expand. At higher doses, expansion of haematopoietic cells is reduced as compared with non‐irradiated cultures but it remains significant. This reduction in expansion of BMMNC was related to radiation‐induced apoptosis. Conclusion: The results suggest that it is possible to expand haematopoietic cells after irradiation doses at least up to 2 Gy. This suggests a possible use of cell therapy for the treatment of radiation accident victims.


Journal of Hematotherapy & Stem Cell Research | 2002

Reinjection of Ex Vivo–Expanded Primate Bone Marrow Mononuclear Cells Strongly Reduces Radiation-Induced Aplasia

Jean-Marc Bertho; Johanna Frick; Christelle Demarquay; Armelle Lauby; Emilie Mathieu; Nicolas Dudoignon; Nelly Jacquet; F. Trompier; Alain Chapel; Christophe Joubert; Manuel Lopez; Jocelyne Aigueperse; Norbert-Claude Gorin; Patrick Gourmelon; Dominique Thierry

To assess the therapeutic efficacy of ex vivo-expanded hematopoietic cells in the treatment of radiation-induced pancytopenia, we have set up a non-human primate model. Two ex vivo expansion protocols for bone marrow mononuclear cells (BMMNC) were studied. The first consisted of a 7-day culture in the presence of stem cell factor (SCF), Flt3-ligand, thrombopoietin (TPO), interleukin-3 (IL-3), and IL-6, which induced preferentially the expansion of immature hematopoietic cells [3.1 +/- 1.4, 10.0 +/- 5.1, 2.2 +/- 1.9, and 1.0 +/- 0.3-fold expansion for mononuclear cells (MNC), colony-forming units-granulocyte-macrophage (CFU-GM), burst-forming units erythroid (BFU-E), and long-term culture initiating cells (LTC-IC) respectively]. The second was with the same cytokine combination supplemented with granulocyte colony-stimulating factor (G-CSF) with an increased duration of culture up to 14 days and induced mainly the production of mature hematopoietic cells (17.2 +/- 11.7-fold expansion for MNC and no detectable BFU-E and LTC-IC), although expansion of CFU-GM (13.7 +/- 18.8-fold) and CD34+ cells (5.2 +/- 1.4-fold) was also observed. Results showed the presence of mesenchymal stem cells and cells from the lymphoid and the megakaryocytic lineages in 7-day expanded BMMNC. To test the ability of ex vivo-expanded cells to sustain hematopoietic recovery after radiation-induced aplasia, non-human primates were irradiated at a supralethal dose of 8 Gy and received the product of either 7-day (24 h after irradiation) or 14-day (8 days after irradiation) expanded BMMNC. Results showed that the 7-day ex vivo-expanded BMMNC shortened the period and the severity of pancytopenia and improved hematopoietic recovery, while the 14 day ex vivo-expanded BMMNC mainly produced a transfusion-like effect during 8 days, followed by hematopoietic recovery. These results suggest that ex vivo expanded BMMNC during 7 days may be highly efficient in the treatment of radiation-induced aplasia.


International Journal of Radiation Biology | 2006

Follow-up of stable chromosomal aberrations in gamma-ray irradiated non-human primates

Eric Gregoire; Irène Sorokine-Durm; Jean-Marc Bertho; N. Jacquet; M. Delbos; Christelle Demarquay; Philippe Voisin; Laurence Roy

Purpose: The purpose of this study was to examine a new approach to retrospective biological dosimetry, by using a long-term animal model to determine the stability of translocation frequency after in vivo irradiation. While the frequency of dicentrics is known to decrease over time, the persistence of more stable chromosomal aberrations such as translocations could be useful if their stability were definitively proved. Materials and methods: Four monkeys (Macaca fascicularis) were exposed to two different doses of ionizing radiation: 2 Gy whole body irradiation for two and 4 Gy for two others. Blood samples were obtained at various times after irradiation. Both total and two-way translocations were detected by fluorescence in situ hybridization. Translocations were scored in stable cells, that is, those without dicentrics, rings or fragments. The course of translocation frequency was analysed at four time-points: one hour (H1), 2 months (M2), 10 months (M10) and 31 months (M31) after irradiation. Results: We observed two separate trends in translocation frequency: Total translocation frequency decreased slightly in animals irradiated with a dose of 2 Gy, while two-way translocation frequency was relatively stable in all irradiated animals. Conclusions: We confirmed the long-term stability of translocations and found that it seems to depend on the type of the translocation recorded. Overall translocations were stable for up to 31 months regardless of dose, but two-way translocations were more stable than those that were non-reciprocal, especially in stable cells.


The Journal of Pathology | 2015

TH17 predominant T-cell responses in radiation-induced bowel disease are modulated by treatment with adipose-derived mesenchymal stromal cells.

Raphaëlle Bessout; Christelle Demarquay; Lara Moussa; Alice René; Bastien Doix; Marc Benderitter; Alexandra Sémont; Noëlle Mathieu

Radiation proctitis is an insidious disease associated with substantial morbidity and mortality. It may develop following the treatment of several cancers by radiotherapy when normal colorectal tissues are present in the irradiation field. There is no unified approach for the assessment and treatment of this disease, partly due to insufficient knowledge about the mechanism involved in the development of radiation proctitis. However, unresolved inflammation is hypothesized to have an important role in late side effects. This study aimed to analyse the involvement of specific immunity in colorectal damage developing after localized irradiation, and evaluate the benefit of immunomodulatory mesenchymal stromal cells isolated from adipose tissue (Ad‐MSCs) for reduction of late side effects. Our experimental model of colorectal irradiation induced severe colonic mucosal damage and fibrosis that was associated with T‐cell infiltration. Immune cell activation was investigated; adoptive transfer of T cells in nude rats showed stronger colonization by T cells isolated from irradiated rats. The predominant role of T cells in late radiation‐induced damage and regeneration processes was highlighted by in vivo depletion experiments. Treatments using Ad‐MSCs reduced T‐cell infiltration in the colon and reduced established colonic damage as measured by histological score, functional circular muscle contractibility, and collagen deposition. Here, we have demonstrated for the first time the predominance of the TH17 population compared to TH1 and TH2 in radiation‐induced bowel disease, and that this is reduced after Ad‐MSC treatment. Additionally, we demonstrated in vitro that IL17 acts directly on colonic smooth muscle cells to induce expression of pro‐inflammatory genes that could participate in the development of radiation‐induced injury. Our data demonstrate that the TH17 population is specifically induced during development of radiation‐induced side effects in the colon. Moreover, Ad‐MSC treatment modulates the TH17 population and reduces the extracellular matrix remodelling process induced following irradiation. Copyright

Collaboration


Dive into the Christelle Demarquay's collaboration.

Top Co-Authors

Avatar

Patrick Gourmelon

Institut de radioprotection et de sûreté nucléaire

View shared research outputs
Top Co-Authors

Avatar

Dominique Thierry

Institut de radioprotection et de sûreté nucléaire

View shared research outputs
Top Co-Authors

Avatar

Johanna Frick

Institut de radioprotection et de sûreté nucléaire

View shared research outputs
Top Co-Authors

Avatar

Noëlle Mathieu

Institut de radioprotection et de sûreté nucléaire

View shared research outputs
Top Co-Authors

Avatar

Jean Marc Bertho

Institut de radioprotection et de sûreté nucléaire

View shared research outputs
Top Co-Authors

Avatar

Jean-Marc Bertho

Institut de radioprotection et de sûreté nucléaire

View shared research outputs
Top Co-Authors

Avatar

Alain Chapel

Institut de radioprotection et de sûreté nucléaire

View shared research outputs
Top Co-Authors

Avatar

Alexandra Sémont

Institut de radioprotection et de sûreté nucléaire

View shared research outputs
Top Co-Authors

Avatar

Marc Benderitter

Institut de radioprotection et de sûreté nucléaire

View shared research outputs
Top Co-Authors

Avatar

Lara Moussa

Institut de radioprotection et de sûreté nucléaire

View shared research outputs
Researchain Logo
Decentralizing Knowledge