Network


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

Hotspot


Dive into the research topics where Alain Chapel is active.

Publication


Featured researches published by Alain Chapel.


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.


Stem Cells | 2006

Local irradiation not only induces homing of human mesenchymal stem cells at exposed sites but promotes their widespread engraftment to multiple organs : A study of their quantitative distribution after irradiation damage

Sabine Francois; Morad Bensidhoum; Moubarak Mouiseddine; Christelle Mazurier; Bénédicte Allenet; Alexandra Sémont; Johanna Frick; Amandine Saché; Sandrine Bouchet; Dominique Thierry; Patrick Gourmelon; Gorin Nc; Alain Chapel

Mesenchymal stem cells (MSCs) have been shown to migrate to various tissues. There is little information on the fate and potential therapeutic efficacy of the reinfusion of MSCs following total body irradiation (TBI). We addressed this question using human MSC (hMSCs) infused to nonobese diabetic/ severe combined immunodeficient (NOD/SCID) mice submitted to TBI. Further, we tested the impact of additional local irradiation (ALI) superimposed to TBI, as a model of accidental irradiation. NOD/SCID mice were transplanted with hM‐SCs. Group 1 was not irradiated before receiving hMSC infusion. Group 2 received only TBI at a dose of 3.5 Gy, group 3 received local irradiation to the abdomen at a dose of 4.5 Gy in addition to TBI, and group 4 received local irradiation to the leg at 26.5 Gy in addition to TBI. Fifteen days after irradiation, quantitative and spatial distribution of the hMSCs were studied. Histological analysis of mouse tissues confirmed the presence of radio‐induced lesions in the irradiated fields. Following their infusion into nonirradiated animals, hMSCs homed at a very low level to various tissues (lung, bone marrow, and muscles) and no significant engraftment was found in other organs. TBI induced an increase of engraftment levels of hMSCs in the brain, heart, bone marrow, and muscles. Abdominal irradiation (AI) as compared with leg irradiation (LI) increased hMSC engraftment in the exposed area (the gut, liver, and spleen). Hind LI as compared with AI increased hMSC engraftment in the exposed area (skin, quadriceps, and muscles). An increase of hMSC engraftment in organs outside the fields of the ALI was also observed. Conversely, following LI, hMSC engraftment was increased in the brain as compared with AI. This study shows that engraftment of hMSCs in NOD/ SCID mice with significantly increased in response to tissue injuries following TBI with or without ALI. ALI induced an increase of the level of engraftment at sites outside the local irradiation field, thus suggesting a distant (abscopal) effect of radiation damage. This work supports the use of MSCs to repair damaged normal tissues following accidental irradiation and possibly in patients submitted to radiotherapy.


Advances in Experimental Medicine and Biology | 2006

Mesenchymal Stem Cells Increase Self-Renewal of Small Intestinal Epithelium and Accelerate Structural Recovery after Radiation Injury

Alexandra Sémont; Sabine Francois; Moubarak Mouiseddine; Agnès François; Amandine Saché; Johanna Frick; Dominique Thierry; Alain Chapel

Patients who undergo pelvic or abdominal radiotherapy may develop side effects that can be life threatening. Tissue complications caused by radiation-induced stem cell depletion may result in structural and functional alterations of the gastrointestinal (GI) tract. Stem cell therapy using mesenchymal stem cells (MSC) is a promising approach for replenishment of the depleted stem cell compartment during radiotherapy. There is little information on the therapeutic potential of MSC in injured-GI tract following radiation exposure. In this study, we addressed the ability of MSC to support the structural regeneration of the small intestine after abdominal irradiation. We isolated MSC from human bone marrow and human mesenchymal stem cells (hMSC) were transplanted into immunotolerent NOD/SCID mice with a dose of 5.10(6) cells via the systemic route. Using a model of radiation-induced intestinal injury, we studied the link between damage, hMSC engraftment and the capacity of hMSC to sustain structural recovery. Tissue injury was assessed by histological analysis. hMSC engraftment in tissues was quantified by PCR assay. Following abdominal irradiation, the histological analysis of small intestinal structure confirms the presence of partial and transient (three days) mucosal atrophy. PCR analysis evidences a low but significant hMSC implantation in small intestine (0.17%) but also at all the sites of local irradiation (kidney, stomach and spleen). Finally, in presence of hMSC, the small intestinal structure is already recovered at three days after abdominal radiation exposure. We show a structural recovery accompanied by an increase of small intestinal villus height, three and fifteen days following abdominal radiation exposure. In this study, we show that radiation-induced small intestinal injury may play a role in the recruitment of MSC for the improvement of tissue recovery. This work supports, the use of MSC infusion to repair damaged GI tract in patients subjected to radiotherapy. MSC therapy to avoid extended intestinal crypt sterilization is a promising approach to diminish healthy tissue alterations during the course of pelvic radiotherapy.


Clinical Reviews in Allergy & Immunology | 2013

Use of Mesenchymal Stem Cells (MSC) in Chronic Inflammatory Fistulizing and Fibrotic Diseases: a Comprehensive Review

Jan Voswinkel; Sabine Francois; Jean-Marc Simon; Marc Benderitter; Norbert-Claude Gorin; Mohamad Mohty; Loic Fouillard; Alain Chapel

Mesenchymal stem cells (MSC), multipotent adult stem cells, feature the potential to regenerate tissue damage and, in parallel, inhibit inflammation and fibrosis. MSC can be safely transplanted in autologous and allogeneic ways as they are non-immunogenic, and consequently represent a therapeutic option for refractory connective tissue diseases, fibrosing diseases like scleroderma and fistulizing colitis like in Crohn’s disease. Actually, there are more than 200 registered clinical trial sites for evaluating MSC therapy, and 22 are on autoimmune diseases. In irradiation-induced colitis, MSC accelerate functional recovery of the intestine and dampen the systemic inflammatory response. In order to provide rescue therapy for accidentally over-irradiated prostate cancer patients who underwent radiotherapy, allogeneic bone marrow-derived MSC from family donors were intravenously infused to three patients with refractory and fistulizing colitis resembling fistulizing Crohn’s disease. Systemic MSC therapy of refractory irradiation-induced colitis was safe and effective on pain, diarrhoea, hemorrhage, inflammation and fistulization accompanied by modulation of the lymphocyte subsets towards an increase of T regulatory cells and a decrease of activated effector T cells. The current data indicate that MSC represent a promising alternative strategy in the treatment of various immune-mediated diseases. Encouraging results have already been obtained from clinical trials in Crohn’s disease and SLE as well as from case series in systemic sclerosis. MSC represent a safe therapeutic measure for patients who suffer from chronic and fistulizing colitis. These findings are instructional for the management of refractory inflammatory bowel diseases that are characterized by similar clinical and immunopathological features.


Haematologica | 2012

Human induced pluripotent stem cells can reach complete terminal maturation: in vivo and in vitro evidence in the erythropoietic differentiation model

Ladan Kobari; Frank Yates; Noufissa Oudrhiri; Alain Francina; Laurent Kiger; Christelle Mazurier; Wassim El-Nemer; Nicolas Hebert; Marie-Catherine Giarratana; Sabine Francois; Alain Chapel; Hélène Lapillonne; Dominique Luton; Annelise Bennaceur-Griscelli; Luc Douay

Background Human induced pluripotent stem cells offer perspectives for cell therapy and research models for diseases. We applied this approach to the normal and pathological erythroid differentiation model by establishing induced pluripotent stem cells from normal and homozygous sickle cell disease donors. Design and Methods We addressed the question as to whether these cells can reach complete erythroid terminal maturation notably with a complete switch from fetal to adult hemoglobin. Sickle cell disease induced pluripotent stem cells were differentiated in vitro into red blood cells and characterized for their terminal maturation in terms of hemoglobin content, oxygen transport capacity, deformability, sickling and adherence. Nucleated erythroblast populations generated from normal and pathological induced pluripotent stem cells were then injected into non-obese diabetic severe combined immunodeficiency mice to follow the in vivo hemoglobin maturation. Results We observed that in vitro erythroid differentiation results in predominance of fetal hemoglobin which rescues the functionality of red blood cells in the pathological model of sickle cell disease. We observed, in vivo, the switch from fetal to adult hemoglobin after infusion of nucleated erythroid precursors derived from either normal or pathological induced pluripotent stem cells into mice. Conclusions These results demonstrate that human induced pluripotent stem cells: i) can achieve complete terminal erythroid maturation, in vitro in terms of nucleus expulsion and in vivo in terms of hemoglobin maturation; and ii) open the way to generation of functionally corrected red blood cells from sickle cell disease induced pluripotent stem cells, without any genetic modification or drug treatment.


Stem Cells International | 2014

Management of Fibrosis: The Mesenchymal Stromal Cells Breakthrough

Benoît Usunier; Marc Benderitter; Radia Tamarat; Alain Chapel

Fibrosis is the endpoint of many chronic inflammatory diseases and is defined by an abnormal accumulation of extracellular matrix components. Despite its slow progression, it leads to organ malfunction. Fibrosis can affect almost any tissue. Due to its high frequency, in particular in the heart, lungs, liver, and kidneys, many studies have been conducted to find satisfactory treatments. Despite these efforts, current fibrosis management therapies either are insufficiently effective or induce severe adverse effects. In the light of these facts, innovative experimental therapies are being investigated. Among these, cell therapy is regarded as one of the best candidates. In particular, mesenchymal stromal cells (MSCs) have great potential in the treatment of inflammatory diseases. The value of their immunomodulatory effects and their ability to act on profibrotic factors such as oxidative stress, hypoxia, and the transforming growth factor-β1 pathway has already been highlighted in preclinical and clinical studies. Furthermore, their propensity to act depending on the microenvironment surrounding them enhances their curative properties. In this paper, we review a large range of studies addressing the use of MSCs in the treatment of fibrotic diseases. The results reported here suggest that MSCs have antifibrotic potential for several organs.


Oncogene | 2005

Chemosensitization by erythropoietin through inhibition of the NF-kappaB rescue pathway

Gabrielle Carvalho; Carmen Lefaucheur; Claire Cherbonnier; Didier Métivier; Alain Chapel; Marc Pallardy; Marie-Françoise Bourgeade; Bernard Charpentier; François Hirsch; Guido Kroemer

Two cell lines that exemplify erythropoietin (EPO) receptor-positive tumors, human renal carcinoma cell lines RCC and the myelomonocytic leukemia cell line U937, were investigated for the apoptosis-modulatory potential of EPO. Cells cultured in the presence of EPO exhibited an elevated apoptotic response to cancer chemotherapeutic agents such as daunorubicin (Dauno) and vinblastine (VBL). Chemosensitization by EPO did not involve an increase in p53 activation, yet correlated with enhanced Bax/Bak-dependent mitochondrial membrane perturbation and caspase maturation. In vitro monotherapy with Dauno or VBL induced the degradation of IκBα, provoked the translocation of NF-κB p65/50 to the nucleus and stimulated the expression of an NF-κB-activatable reporter gene. All these signs of NF-κB activation were perturbed in the presence of EPO. Inhibition of JAK2, one of the receptor-proximal elements of EPO-mediated signal transduction, greatly diminished the EPO-mediated chemosensitization and NF-κB inhibition. EPO lost its death-facilitating effects in the presence of an NF-κB inhibitor, underscoring the cause–effect relationship between EPO-mediated chemosensitization and NF-κB inhibition. Altogether, these results suggest that, at least in a specific subset of tumors, EPO receptor agonists can prevent activation of the NF-κB pathway, thereby enhancing the propensity of EPO receptor-positive tumor cells to undergo apoptosis.


Health Physics | 2010

New emerging concepts in the medical management of local radiation injury.

Marc Benderitter; Patrick Gourmelon; Eric Bey; Alain Chapel; I. Clairand; Marie Prat; Jean Jacques Lataillade

Treatment of severe radiation burns remains a difficult medical challenge. The response of the skin to ionizing radiation results in a range of clinical manifestations. The most severe manifestations are highly invalidating. Although several therapeutic strategies (excision, skin grafting, skin or muscle flaps) have been used with some success, none have proven entirely satisfying. The concept that stem cell injections could be used for reducing normal tissue injury has been discussed for a number of years. Mesenchymal stem cells therapy may be a promising therapeutic approach for improving radiation-induced skin and muscle damages. Pre-clinical and clinical benefit of mesenchymal stem cell injection for ulcerated skin and muscle restoration after high dose radiation exposure has been successfully demonstrated. Three first patients suffering from severe radiological syndrome were successfully treated in France based on autologous human grade mesenchymal stem cell injection combined to plastic surgery or skin graft. Stem cell therapy has to be improved to the point that hospitals can put safe, efficient, and reliable clinical protocols into practice.


International Journal of Radiation Oncology Biology Physics | 2003

PLASMA Flt-3 LIGAND CONCENTRATION CORRELATED WITH RADIATION-INDUCED BONE MARROW DAMAGE DURING LOCAL FRACTIONATED RADIOTHERAPY

Aymeri Huchet; Yazid Belkacemi; Johanna Frick; Marie Prat; Ioanna Muresan-Kloos; Dan Altan; Alain Chapel; Norbert Claude Gorin; Patrick Gourmelon; Jean Marc Bertho

PURPOSE To determine whether variations in the plasma Flt-3 ligand (FL) concentration after radiotherapy (RT) may serve as a biomarker for radiation-induced bone marrow damage. METHODS AND MATERIALS Twenty-seven patients were followed during RT. The irradiated bone marrow volume was determined. The blood cell counts and plasma FL concentrations were evaluated before and after RT. The expression of membrane-bound FL and mRNA expression were also defined in circulating blood cells. RESULTS We found a negative correlation between the plasma FL concentration and the number of circulating white blood cells and platelets during RT. Moreover, the overall amount of FL in the blood of patients during RT correlated directly with both the cumulated radiation dose and the proportion of irradiated bone marrow. CONCLUSIONS We demonstrated that the variations in plasma FL concentration directly reflect the radiation-induced bone marrow damage during fractionated local RT. We suggest a possible use for FL monitoring as a means to predict the occurrence of Grade 3-4 leukopenia or thrombocytopenia during the course of RT.


BioMed Research International | 2013

Human Mesenchymal Stem Cells Provide Protection against Radiation-Induced Liver Injury by Antioxidative Process, Vasculature Protection, Hepatocyte Differentiation, and Trophic Effects

Sabine Francois; Moubarak Mouiseddine; Bénédicte Allenet-Lepage; Jan Voswinkel; Luc Douay; Marc Benderitter; Alain Chapel

To evaluate the potential therapeutic effect of the infusion of hMSCs for the correction of liver injuries, we performed total body radiation exposure of NOD/SCID mice. After irradiation, mir-27b level decreases in liver, increasing the directional migration of hMSCs by upregulating SDF1α. A significant increase in plasmatic transaminases levels, apoptosis process in the liver vascular system, and in oxidative stress were observed. hMSC injection induced a decrease in transaminases levels and oxidative stress, a disappearance of apoptotic cells, and an increase in Nrf2, SOD gene expression, which might reduce ROS production in the injured liver. Engrafted hMSCs expressed cytokeratin CK18 and CK19 and AFP genes indicating possible hepatocyte differentiation. The presence of hMSCs expressing VEGF and Ang-1 in the perivascular region, associated with an increased expression of VEGFr1, r2 in the liver, can confer a role of secreting cells to hMSCs in order to maintain the endothelial function. To explain the benefits to the liver of hMSC engraftment, we find that hMSCs secreted NGF, HGF, and anti-inflammatory molecules IL-10, IL1-RA contributing to prevention of apoptosis, increasing cell proliferation in the liver which might correct liver dysfunction. MSCs are potent candidates to repair and protect healthy tissues against radiation damages.

Collaboration


Dive into the Alain Chapel's collaboration.

Top Co-Authors

Avatar

Sabine Francois

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

Patrick Gourmelon

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

Moubarak Mouiseddine

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

Johanna Frick

Institut de radioprotection et de sûreté nucléaire

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Christelle Demarquay

Institut de radioprotection et de sûreté nucléaire

View shared research outputs
Top Co-Authors

Avatar

Amandine Saché

Institut de radioprotection et de sûreté nucléaire

View shared research outputs
Researchain Logo
Decentralizing Knowledge