Network


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

Hotspot


Dive into the research topics where Danièle Noël is active.

Publication


Featured researches published by Danièle Noël.


Stem Cells | 2007

Mesenchymal Stem Cells Inhibit the Differentiation of Dendritic Cells Through an Interleukin‐6‐Dependent Mechanism

Farida Djouad; Louis-Marie Charbonnier; Carine Bouffi; Pascale Louis-Plence; Claire Bony; Florence Apparailly; Céline Cantos; Christian Jorgensen; Danièle Noël

Mesenchymal stem cells (MSC) are of particular interest for their potential clinical use in tissue engineering as well as for their capacity to reduce the incidence and severity of graft‐versus‐host disease in allogeneic transplantation. We have previously shown that MSC‐mediated immune suppression acts via the secretion of soluble factor(s) induced upon stimulation. The aim of this study was to identify the molecule(s) involved and the underlying mechanism(s). We show that murine MSC secrete high levels of interleukin (IL)‐6 and vascular endothelial growth factor, which are directly correlated to the inhibition of T‐cell proliferation. The T‐cell activation is partially restored upon addition of a neutralizing anti‐IL‐6 antibody or the prostaglandin E2 inhibitor indomethacin. Interestingly, no indoleamine 2,3‐dioxygenase activity was detected in our conditions. Instead, we show that MSC reduce the expression of major histocompatibility complex class II, CD40, and CD86 costimulatory molecules on mature dendritic cells (DC), which was responsible for a decrease in T‐cell proliferation. Moreover, we show that the differentiation of bone marrow progenitors into DC cultured with conditioned supernatants from MSC was partly inhibited through the secretion of IL‐6. Altogether, these data suggest that IL‐6 is involved in the immunoregulatory mechanism mediated by MSC through a partial inhibition of DC differentiation but is probably not the main mechanism.


Trends in Biotechnology | 2009

Cartilage engineering: a crucial combination of cells, biomaterials and biofactors

Claire Vinatier; Dominique Mrugala; Christian Jorgensen; Jérôme Guicheux; Danièle Noël

Injuries to articular cartilage are one of the most challenging issues of musculoskeletal medicine due to the poor intrinsic ability of this tissue for repair. The lack of efficient modalities of treatment has prompted research into tissue engineering combining chondrogenic cells, scaffold materials and environmental factors. The aim of this review is to focus on the recent advances made in exploiting the potential of biomaterial-assisted cell therapy for cartilage engineering. We discuss the requirements for identifying additional specific growth factors and evaluating the optimal combination of cells, growth factors and scaffolds that is able to respond to the functional demand placed upon cartilage tissue replacement in clinics. Finally, some of the major obstacles encountered in cartilage engineering are discussed, as well as future trends in clinical applications.


Stem Cell Research & Therapy | 2010

Immunosuppression by mesenchymal stem cells: mechanisms and clinical applications

Soufiane Ghannam; Carine Bouffi; Farida Djouad; Christian Jorgensen; Danièle Noël

Mesenchymal stem cells (MSCs) are multipotential nonhematopoietic progenitor cells that are isolated from many adult tissues, in particular from the bone marrow and adipose tissue. Along with their capacity for differentiating into cells of mesodermal lineage, such as adipocytes, osteoblasts and chondrocytes, these cells have also generated great interest for their ability to display immunomodulatory capacities. Indeed, a major breakthrough came with the finding that they are able to induce peripheral tolerance, suggesting they may be used as therapeutic tools in immune-mediated disorders. The present review aims at discussing the current knowledge on the targets and mechanisms of MSC-mediated immunosuppression as well as the potential use of MSCs as modulators of immune responses in a variety of diseases related to alloreactive immunity or autoimmunity


Experimental Cell Research | 2008

Cell specific differences between human adipose-derived and mesenchymal–stromal cells despite similar differentiation potentials

Danièle Noël; David Caton; Stéphane Roche; Claire Bony; Sylvain Lehmann; Louis Casteilla; Christian Jorgensen; Béatrice Cousin

Stromal cells from bone marrow and adipose tissue are attractive sources of adult progenitors for cell-based therapy. However, whether those cell populations represent intrinsically different cell types is still largely under debate. The aim of this study was to systematically and quantitatively compare adipose-derived stromal cells (ADSC) and bone marrow-derived multipotent mesenchymal-stromal cells (BM-MSC). The quantitative comparison was realized using Taqman Low Density Array, 2D electrophoresis and differentiation functional assays in vitro. Furthermore, cells engineered to express TGFbeta1 were injected into the intra-articular space of mouse knee joints in order to determine whether they were able to form new differentiated tissues in vivo. Our data revealed cell specific differences at transcriptional and proteomic levels between both cell types according to their tissue origin as well as functional differences in their differentiation processes towards adipogenic, osteogenic and chondrogenic programs. Nevertheless, in vitro as well as in vivo ADSC displayed the same ability than MSC to differentiate towards chondrocytes/osteoblasts, comforting the status of both cell sources as promising regenerative cells. In summary, our observations indicate that ADSC and MSC are fundamentally different cell types and differently committed cells.


PLOS ONE | 2010

IL-6-Dependent PGE2 Secretion by Mesenchymal Stem Cells Inhibits Local Inflammation in Experimental Arthritis

Carine Bouffi; Claire Bony; Gabriel Courties; Christian Jorgensen; Danièle Noël

Background Based on their capacity to suppress immune responses, multipotent mesenchymal stromal cells (MSC) are intensively studied for various clinical applications. Although it has been shown in vitro that the immunomodulatory effect of MSCs mainly occurs through the secretion of soluble mediators, the mechanism is still not completely understood. The aim of the present study was to better understand the mechanisms underlying the suppressive effect of MSCs in vivo, using cells isolated from mice deficient in the production of inducible nitric oxide synthase (iNOS) or interleukin (IL)-6 in the murine model of collagen-induced arthritis. Principal Findings In the present study, we show that primary murine MSCs from various strains of mice or isolated from mice deficient for iNOS or IL-6 exhibit different immunosuppressive potential. The immunomodulatory function of MSCs was mainly attributed to IL-6-dependent secretion of prostaglandin E2 (PGE2) with a minor role for NO. To address the role of these molecules in vivo, we used the collagen-induced arthritis as an experimental model of immune-mediated disorder. MSCs effectively inhibited collagen-induced inflammation during a narrow therapeutic window. In contrast to wild type MSCs, IL-6-deficient MSCs and to a lesser extent iNOS-deficient MSCs were not able to reduce the clinical signs of arthritis. Finally, we show that, independently of NO or IL-6 secretion or Treg cell induction, MSCs modulate the host response by inducing a switch to a Th2 immune response. Significance Our data indicate that MSCs mediate their immunosuppressive effect via two modes of action: locally, they reduce inflammation through the secretion of anti-proliferative mediators, such as NO and mainly PGE2, and systemically they switch the host response from a Th1/Th17 towards a Th2 immune profile.


Stem Cells | 2004

Short-Term BMP-2 Expression Is Sufficient for In Vivo Osteochondral Differentiation of Mesenchymal Stem Cells

Danièle Noël; Dan Gazit; Céline Bouquet; Florence Apparailly; Claire Bony; Pascale Plence; Virginie Millet; Gadi Turgeman; Michel Perricaudet; Sany J; Christian Jorgensen

Currently available murine models to evaluate mesenchymal stem cell (MSC) differentiation are based on cell injection at ectopic sites such as muscle or skin. Due to the importance of environmental factors on the differentiation capacities of stem cells in vivo, we investigated whether the peculiar synovial/cartilaginous environment may influence the lineage specificity of bone morphogenetic protein (BMP)‐2‐engineered MSCs. To this aim, we used the C3H10T1/2‐derived C9 MSCs that express BMP‐2 under control of the doxycycline (Dox)‐repressible promoter, Tet‐Off, and showed in vitro, using the micropellet culture system that C9 MSCs kept their potential to differentiate toward chondrocytes. Implantation of C9 cells, either into the tibialis anterior muscles or into the joints of CB17‐severe combined immunodeficient bg mice led to the formation of cartilage and bone filled with bone marrow as soon as day 10. However, no differentiation was observed after injection of naïve MSCs or C9 cells that were repressed to secrete BMP‐2 by Dox addition. The BMP‐2‐induced differentiation of adult MSCs is thus independent of soluble factors present in the local environment of the synovial/cartilaginous tissues. Importantly, we demonstrated that a short‐term expression of the BMP‐2 growth factor is necessary and sufficient to irreversibly induce bone formation, suggesting that a stable genetic modification of MSCs is not required for stem cell‐based bone/cartilage engineering.


Nature Reviews Rheumatology | 2009

Mesenchymal stem cells: innovative therapeutic tools for rheumatic diseases

Farida Djouad; Carine Bouffi; Soufiane Ghannam; Danièle Noël; Christian Jorgensen

Mesenchymal stem cells (MSCs), or multipotent mesenchymal stromal cells as they are also known, have been identified in bone marrow as well as in other tissues of the joint, including adipose, synovium, periosteum, perichondrium, and cartilage. These cells are characterized by their phenotype and their ability to differentiate into three lineages: chondrocytes, osteoblasts and adipocytes. Importantly, MSCs also potently modulate immune responses, exhibit healing capacities, improve angiogenesis and prevent fibrosis. These properties might be explained at least in part by the trophic effects of MSCs through the secretion of a number of cytokines and growth factors. However, the mechanisms involved in the differentiation potential of MSCs, and their immunomodulatory and paracrine properties, are currently being extensively studied. These unique properties of MSCs confer on them the potential to be used for therapeutic applications in rheumatic diseases, including rheumatoid arthritis, osteoarthritis, genetic bone and cartilage disorders as well as bone metastasis.


Stem Cells | 2006

Functional Neuronal Differentiation of Bone Marrow‐Derived Mesenchymal Stem Cells

Philippe Tropel; Nadine Platet; Jean-Claude Platel; Danièle Noël; Mireille Albrieux; Alim-Louis Benabid; François Berger

Recent results have shown the ability of bone marrow cells to migrate in the brain and to acquire neuronal or glial characteristics. In vitro, bone marrow‐derived MSCs can be induced by chemical compounds to express markers of these lineages. In an effort to set up a mouse model of such differentiation, we addressed the neuronal potentiality of mouse MSCs (mMSCs) that we recently purified. These cells expressed nestin, a specific marker of neural progenitors. Under differentiating conditions, mMSCs display a distinct neuronal shape and express neuronal markers NF‐L (neurofilament‐light, or neurofilament 70 kDa) and class III β‐tubulin. Moreover, differentiated mMSCs acquire neuron‐like functions characterized by a cytosolic calcium rise in response to various specific neuronal activators. Finally, we further demonstrated for the first time that clonal mMSCs and their progeny are competent to differentiate along the neuronal pathway, demonstrating that these bone marrow‐derived stem cells share characteristics of widely multipotent stem cells unrestricted to mesenchymal differentiation pathways.


Arthritis Research & Therapy | 2005

Transcriptional profiles discriminate bone marrow-derived and synovium-derived mesenchymal stem cells

Farida Djouad; Claire Bony; Thomas Häupl; Gilles Uzé; Najiba Lahlou; Pascale Louis-Plence; Florence Apparailly; François Canovas; Thierry Rème; Sany J; Christian Jorgensen; Danièle Noël

Previous studies have reported that mesenchymal stem cells (MSC) may be isolated from the synovial membrane by the same protocol as that used for synovial fibroblast cultivation, suggesting that MSC correspond to a subset of the adherent cell population, as MSC from the stromal compartment of the bone marrow (BM). The aims of the present study were, first, to better characterize the MSC derived from the synovial membrane and, second, to compare systematically, in parallel, the MSC-containing cell populations isolated from BM and those derived from the synovium, using quantitative assays. Fluorescent-activated cell sorting analysis revealed that both populations were negative for CD14, CD34 and CD45 expression and that both displayed equal levels of CD44, CD73, CD90 and CD105, a phenotype currently known to be characteristic of BM-MSC. Comparable with BM-MSC, such MSC-like cells isolated from the synovial membrane were shown for the first time to suppress the T-cell response in a mixed lymphocyte reaction, and to express the enzyme indoleamine 2,3-dioxygenase activity to the same extent as BM-MSC, which is a possible mediator of this suppressive activity. Using quantitative RT-PCR these data show that MSC-like cells from the synovium and BM may be induced to chondrogenic differentiation and, to a lesser extent, to osteogenic differentiation, but the osteogenic capacities of the synovium-derived MSC were significantly reduced based on the expression of the markers tested (collagen type II and aggrecan or alkaline phosphatase and osteocalcin, respectively). Transcription profiles, determined with the Atlas Human Cytokine/Receptor Array, revealed discrimination between the MSC-like cells from the synovial membrane and the BM-MSC by 46 of 268 genes. In particular, activin A was shown to be one major upregulated factor, highly secreted by BM-MSC. Whether this reflects a different cellular phenotype, a different amount of MSC in the synovium-derived population compared with BM-MSC adherent cell populations or the impact of a different microenvironment remains to be determined. In conclusion, although the BM-derived and synovium-derived MSC shared similar phenotypic and functional properties, both their differentiation capacities and transcriptional profiles permit one to discriminate the cell populations according to their tissue origin.


Stem Cell Research & Therapy | 2013

Mesenchymal stem cells generate a CD4+CD25+Foxp3+ regulatory T cell population during the differentiation process of Th1 and Th17 cells

Patricia Luz-Crawford; Mónica Kurte; Javiera Bravo-Alegría; Rafael Contreras; Estefania Nova-Lamperti; Gautier Tejedor; Danièle Noël; Christian Jorgensen; Fernando Figueroa; Farida Djouad; Flavio Carrión

IntroductionMesenchymal stem cells (MSCs) are adult, multipotent, stem cells with immunomodulatory properties. The mechanisms involved in the capacity of MSCs to inhibit the proliferation of proinflammatory T lymphocytes, which appear responsible for causing autoimmune disease, have yet to be fully elucidated. One of the underlying mechanisms studied recently is the ability of MSCs to generate T regulatory (Treg) cells in vitro and in vivo from activated peripheral blood mononuclear cells (PBMC), T-CD4+ and also T-CD8+ cells. In the present work we investigated the capacity of MSCs to generate Treg cells using T-CD4+ cells induced to differentiate toward the proinflammatory Th1 and Th17 lineages.MethodsMSCs were obtained from mouse bone marrow and characterized according to their surface antigen expression and their multilineage differentiation potential. CD4+ T cells isolated from mouse spleens were induced to differentiate into Th1 or Th17 cells and co-cultured with MSCs added at day 0, 2 or 4 of the differentiation processes. After six days, CD25, Foxp3, IL-17 and IFN-γ expression was assessed by flow cytometry and helios and neuropilin 1 mRNA levels were assessed by RT-qPCR. For the functional assays, the ‘conditioned’ subpopulation generated in the presence of MSCs was cultured with concanavalin A-activated CD4+ T cells labeled with carboxyfluorescein succinimidyl ester. Finally, we used the encephalomyelitis autoimmune diseases (EAE) mouse model, in which mice were injected with MSCs at day 18 and 30 after immunization. At day 50, the mice were euthanized and draining lymph nodes were extracted for Th1, Th17 and Treg detection by flow cytometry.ResultsMSCs were able to suppress the proliferation, activation and differentiation of CD4+ T cells induced to differentiate into Th1 and Th17 cells. This substantial suppressive effect was associated with an increase of the percentage of functional induced CD4+CD25+Foxp3+ regulatory T cells and IL-10 secretion. However, using mature Th1 or Th17 cells our results demonstrated that while MSCs suppress the proliferation and phenotype of mature Th1 and Th17 cells they did not generate Treg cells. Finally, we showed that the beneficial effect observed following MSC injection in an EAE mouse model was associated with the suppression of Th17 cells and an increase in the percentage of CD4+CD25+Foxp3+ T lymphocytes when administrated at early stages of the disease.ConclusionsThis study demonstrated that MSCs contribute to the generation of an immunosuppressive environment via the inhibition of proinflammatory T cells and the induction of T cells with a regulatory phenotype. Together, these results might have important clinical implications for inflammatory and autoimmune diseases.

Collaboration


Dive into the Danièle Noël's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Karine Toupet

University of Montpellier

View shared research outputs
Top Co-Authors

Avatar

Marie Maumus

University of Montpellier

View shared research outputs
Top Co-Authors

Avatar

Farida Djouad

University of Montpellier

View shared research outputs
Top Co-Authors

Avatar

Claire Bony

University of Montpellier

View shared research outputs
Top Co-Authors

Avatar

Marc Piechaczyk

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Carine Bouffi

University of Montpellier

View shared research outputs
Top Co-Authors

Avatar

P. Guilpain

University of Montpellier

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Maxime Ruiz

University of Montpellier

View shared research outputs
Researchain Logo
Decentralizing Knowledge