Network


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

Hotspot


Dive into the research topics where Stéphane D. Girard is active.

Publication


Featured researches published by Stéphane D. Girard.


Journal of Clinical Investigation | 2011

Engraftment of human nasal olfactory stem cells restores neuroplasticity in mice with hippocampal lesions

Emmanuel Nivet; Michel Vignes; Stéphane D. Girard; Caroline Pierrisnard; Nathalie Baril; Arnaud Deveze; Jacques Magnan; Fabien Lanté; Michel Khrestchatisky; François Féron; François S. Roman

Stem cell-based therapy has been proposed as a potential means of treatment for a variety of brain disorders. Because ethical and technical issues have so far limited the clinical translation of research using embryonic/fetal cells and neural tissue, respectively, the search for alternative sources of therapeutic stem cells remains ongoing. Here, we report that upon transplantation into mice with chemically induced hippocampal lesions, human olfactory ecto-mesenchymal stem cells (OE-MSCs) - adult stem cells from human nasal olfactory lamina propria - migrated toward the sites of neural damage, where they differentiated into neurons. Additionally, transplanted OE-MSCs stimulated endogenous neurogenesis, restored synaptic transmission, and enhanced long-term potentiation. Mice that received transplanted OE-MSCs exhibited restoration of learning and memory on behavioral tests compared with lesioned, nontransplanted control mice. Similar results were obtained when OE-MSCs were injected into the cerebrospinal fluid. These data show that OE-MSCs can induce neurogenesis and contribute to restoration of hippocampal neuronal networks via trophic actions. They provide evidence that human olfactory tissue is a conceivable source of nervous system replacement cells. This stem cell subtype may be useful for a broad range of stem cell-related studies.


Methods of Molecular Biology | 2013

Isolation of adult stem cells from the human olfactory mucosa.

François Féron; Chris Perry; Stéphane D. Girard; Alan Mackay-Sim

The olfactory mucosa, located in the nasal cavity, is the only nervous tissue that is exposed to the external environment and easily accessible in every living individual. In addition, this organ is home of a continuing neurogenesis that is sustained by a large population of stem cells. Here, we describe a method for biopsying olfactory mucosa from human nasal cavities and isolating multipotent adult stem cells that can be used to either identify biomarkers in brain disorders or repair the pathological/traumatized nervous system.


Journal of Alzheimer's Disease | 2013

Evidence for early cognitive impairment related to frontal cortex in the 5XFAD mouse model of Alzheimer's disease.

Stéphane D. Girard; Kévin Baranger; Cyrielle Gauthier; Marlyse Jacquet; Anne Bernard; Guy Escoffier; Evelyne Marchetti; Michel Khrestchatisky; Santiago Rivera; François S. Roman

The frontal cortex is a brain structure that plays an important role in cognition and is known to be affected in Alzheimers disease (AD) in humans. Over the past years, transgenic mouse models have been generated to recapitulate the main features of this disease, including cognitive impairments. This study investigates frontal cortex dependent learning abilities in one of the most early-onset transgenic murine model of AD, the 5XFAD mice. We compared frontal performance of 2-, 4-, and 6-month-old 5XFAD mice with their wild-type littermates using a newly developed automated device, the olfactory H-maze, in which mice have to discover three different rules consecutively according to the delayed reaction paradigm. We report early cognitive deficits related to frontal cortex appearing in 4-month-old 5XFAD mice before hippocampal-dependent learning and memory impairment, in relation with neuropathologic processes such as strong gliosis and emerging amyloid plaques. The present results demonstrate that the olfactory H-maze is a very sensitive and simple experimental paradigm that allows assessment of frontal functions in transgenic mice and should be useful to test pre-clinical therapeutic approaches to alter the course of AD.


Cell Transplantation | 2013

Role of matrix metalloproteinases in migration and neurotrophic properties of nasal olfactory stem and ensheathing cells.

Adlane Ould-Yahoui; Oualid Sbai; Kévin Baranger; Anne Bernard; Yatma Gueye; Eliane Charrat; Benoît Clément; Didier Gigmes; Vincent Dive; Stéphane D. Girard; François Féron; Michel Khrestchatisky; Santiago Rivera

Adult olfactory ectomesenchymal stem cells (OE-MSCs) and olfactory ensheathing cells (OECs), both from the nasal olfactory lamina propria, display robust regenerative properties when transplanted into the nervous system, but the mechanisms supporting such therapeutic effects remain unknown. Matrix metalloproteinases (MMPs) are an important family of proteinases contributing to cell motility and axonal outgrowth across the extracellular matrix (ECM) in physiological and pathological conditions. In this study, we have characterized for the first time in nasal human OE-MSCs the expression profile of some MMPs currently associated with cell migration and invasiveness. We demonstrate different patterns of expression for MMP-1, MMP-2, MMP-9, and MT1-MMP upon cell migration when compared with nonmigrating cells. Our results establish a correspondence between the localization of these proteinases in the migration front with the ability of cells to migrate. Using various modulators of MMP activity, we also show that at least MMP-2, MMP-9, and MT1-MMP contribute to OE-MSC migration in an in vitro 3D test. Furthermore, we demonstrate under the same conditions of culture used for in vivo transplantation that OE-MSCs and OECs secrete neurotrophic factors that promote neurite outgrowth of cortical and dorsal root ganglia (DRG) neurons, as well as axo-dendritic differentiation of cortical neurons. These effects were abolished by the depletion of MMP-2 and MMP-9 from the culture conditioned media. Altogether, our results provide the first evidence that MMPs may contribute to the therapeutic features of OE-MSCs and OECs through the control of their motility and/or their neurotrophic properties. Our data provide new insight into the mechanisms of neuroregeneration and will contribute to optimization of cell therapy strategies.


Hippocampus | 2014

Onset of hippocampus-dependent memory impairments in 5XFAD transgenic mouse model of Alzheimer's disease.

Stéphane D. Girard; Marlyse Jacquet; Kévin Baranger; Martine Migliorati; Guy Escoffier; Anne Bernard; Michel Khrestchatisky; François Féron; Santiago Rivera; François S. Roman; Evelyne Marchetti

The 5XFAD mice are an early‐onset transgenic model of Alzheimers disease (AD) in which amyloid plaques are first observed between two and four months of age in the cortical layer five and in the subiculum of the hippocampal formation. Although cognitive alterations have been described in these mice, there are no studies that focused on the onset of hippocampus‐dependent memory deficits, which are a hallmark of the prodromal stage of AD. To identify when the first learning and memory impairments appear, 5XFAD mice of two, four, and six months of age were compared with their respective wild‐type littermates using the olfactory tubing maze, which is a very sensitive hippocampal‐dependent task. Deficits in learning and memory started at four months with a substantial increase at six months of age while no olfactory impairments were observed. The volumetric study using magnetic resonance imaging of the whole brain and specific areas (olfactory bulb, striatum, and hippocampus) did not reveal neuro‐anatomical difference. Slight memory deficits appeared at 4 months of age in correlation with an increased astrogliosis and amyloid plaque formation. This early impairment in learning and memory related to the hippocampal dysfunction is particularly suited to assess preclinical therapeutic strategies aiming to delay or suppress the onset of AD.


Stem Cell Research | 2014

A unique method for the isolation of nasal olfactory stem cells in living rats

Jean-Claude Stamegna; Stéphane D. Girard; Antoine D. Veron; Gilles Sicard; Michel Khrestchatisky; François Féron; François S. Roman

Stem cells are attractive tools to develop new therapeutic strategies for a variety of disorders. While ethical and technical issues, associated with embryonic, fetal and neural stem cells, limit the translation to clinical applications, the nasal stem cells identified in the human olfactory mucosa stand as a promising candidate for stem cell-based therapies. Located in the back of the nose, this multipotent stem cell type is readily accessible in humans, a feature that makes these cells highly suitable for the development of autologous cell-based therapies. However, preclinical studies based on autologous transplantation of rodent olfactory stem cells are impeded because of the narrow opening of the nasal cavity. In this study, we report the development of a unique method permitting to quickly and safely biopsy olfactory mucosa in rats. Using this newly developed technique, rat stem cells expressing the stem cell marker Nestin were successfully isolated without requiring the sacrifice of the donor animal. As an evidence of the self-renewal capacity of the isolated cells, several millions of rat cells were amplified from a single biopsy within four weeks. Using an olfactory discrimination test, we additionally showed that this novel biopsy method does not affect the sense of smell and the learning and memory abilities of the operated animals. This study describes for the first time a methodology allowing the derivation of rat nasal cells in a way that is suitable for studying the effects of autologous transplantation of any cell type present in the olfactory mucosa in a wide variety of rat models.


Frontiers in Molecular Neuroscience | 2017

MT5-MMP Promotes Alzheimer’s Pathogenesis in the Frontal Cortex of 5xFAD Mice and APP Trafficking in vitro

Kévin Baranger; Amandine E. Bonnet; Stéphane D. Girard; Jean-Michel Paumier; Laura García-González; Wejdane Elmanaa; Anne Bernard; Eliane Charrat; Delphine Stephan; Charlotte Bauer; Katrin Moschke; Stefan F. Lichtenthaler; François S. Roman; Frédéric Checler; Michel Khrestchatisky; Santiago Rivera

We previously reported that deficiency of membrane-type five matrix metalloproteinase (MT5-MMP) prevents amyloid pathology in the cortex and hippocampus of 5xFAD mice, and ameliorates the functional outcome. We have now investigated whether the integrity of another important area affected in Alzheimer’s disease (AD), the frontal cortex, was also preserved upon MT5-MMP deficiency in 4-month old mice at prodromal stages of the pathology. We used the olfactory H-maze (OHM) to show that learning impairment associated with dysfunctions of the frontal cortex in 5xFAD was prevented in bigenic 5xFAD/MT5-MMP−/− mice. The latter exhibited concomitant drastic reductions of amyloid beta peptide (Aβ) assemblies (soluble, oligomeric and fibrillary) and its immediate precursor, C99. Simultaneously, astrocyte reactivity and tumor necrosis factor alpha (TNF-α) levels were also lowered. Moreover, MT5-MMP deficiency induced a decrease in N-terminal soluble fragments of amyloid precursor protein (APP), including soluble APPα (sAPPα), sAPPβ and the MT5-MMP-linked fragment of 95 kDa, sAPP95. However, the lack of MT5-MMP did not affect the activity of β- and γ-secretases. In cultured HEKswe cells, transiently expressed MT5-MMP localized to early endosomes and increased the content of APP and Aβ40 in these organelles, as well as Aβ levels in cell supernatants. This is the first evidence that the pro-amyloidogenic features of MT5-MMP lie, at least in part, on the ability of the proteinase to promote trafficking into one of the amyloidogenic subcellular loci. Together, our data further support the pathogenic role of MT5-MMP in AD and that its inhibition improves the functional and pathological outcomes, in this case in the frontal cortex. These data also support the idea that MT5-MMP could become a novel therapeutic target in AD.


Behavioural Brain Research | 2016

The FVB/N mice: A well suited strain to study learning and memory processes using olfactory cues

Stéphane D. Girard; Guy Escoffier; Michel Khrestchatisky; François S. Roman

The FVB/N mice are well suited to generate transgenic animals. These mice are also particularly sensitive to seizures and neurodegeneration induced by systemic administration of chemoconvulsants and are very useful to model epilepsy. However, previous studies report strong cognitive and visual impairments suggesting this background unsuitable for behavioral analysis. In this study, we assessed and compared learning abilities of FVB/N mice to the well characterized C57BL/6 strain using the olfactory tubing maze, a non-visual hippocampus-dependent task in which the mice were trained to learn odor-reward associations. Exploratory behavior and spontaneous locomotor activity were then compared using the open field test. We demonstrated that FVB/N mice were able to learn the task, reaching at the end of the test a high percentage of correct responses. Interestingly, the performance of the FVB/N mice was at least similar to that of the C57BL/6 mice. Moreover, in contrast to previous reports, the FVB/N mice displayed a spontaneous locomotor activity lower than C57BL/6 mice. Our study demonstrated that FVB/N mice are not cognitively impaired and that their learning and memory performance can be assessed when the task is based on olfaction rather than vision.


BMC Veterinary Research | 2018

Isolation and characterization of olfactory ecto-mesenchymal stem cells from eight mammalian genera

Antoine D. Veron; Cécile Bienboire-Frosini; François Féron; Elisa Codecasa; Arnaud Deveze; Dany Royer; Paul Watelet; Pietro Asproni; Kevin Sadelli; Camille Chabaud; Jean-Claude Stamegna; Joël Fagot; Michel Khrestchatisky; Alessandro Cozzi; François S. Roman; Patrick Pageat; Manuel Mengoli; Stéphane D. Girard

BackgroundStem cell-based therapies are an attractive option to promote regeneration and repair defective tissues and organs. Thanks to their multipotency, high proliferation rate and the lack of major ethical limitations, “olfactory ecto-mesenchymal stem cells” (OE-MSCs) have been described as a promising candidate to treat a variety of damaged tissues. Easily accessible in the nasal cavity of most mammals, these cells are highly suitable for autologous cell-based therapies and do not face issues associated with other stem cells. However, their clinical use in humans and animals is limited due to a lack of preclinical studies on autologous transplantation and because no well-established methods currently exist to cultivate these cells. Here we evaluated the feasibility of collecting, purifying and amplifying OE-MSCs from different mammalian genera with the goal of promoting their interest in veterinary regenerative medicine.Biopsies of olfactory mucosa from eight mammalian genera (mouse, rat, rabbit, sheep, dog, horse, gray mouse lemur and macaque) were collected, using techniques derived from those previously used in humans and rats. The possibility of amplifying these cells and their stemness features and differentiation capability were then evaluated.ResultsBiopsies were successfully performed on olfactory mucosa without requiring the sacrifice of the donor animal, except mice. Cell populations were rapidly generated from olfactory mucosa explants. These cells displayed similar key features of their human counterparts: a fibroblastic morphology, a robust expression of nestin, an ability to form spheres and similar expression of surface markers (CD44, CD73). Moreover, most of them also exhibited high proliferation rates and clonogenicity with genus-specific properties. Finally, OE-MSCs also showed the ability to differentiate into mesodermal lineages.ConclusionsThis article describes for the first time how millions of OE-MSCs can be quickly and easily obtained from different mammalian genera through protocols that are well-suited for autologous transplantations. Moreover, their multipotency makes them relevant to evaluate therapeutic application in a wide variety of tissue injury models. This study paves the way for the development of new fundamental and clinical studies based on OE-MSCs transplantation and suggests their interest in veterinary medicine.


Neurobiology of Learning and Memory | 2017

Global cerebral ischemia in rats leads to amnesia due to selective neuronal death followed by astroglial scar formation in the CA1 layer

Kevin Sadelli; Jean-Claude Stamegna; Stéphane D. Girard; Nathalie Baril; Guy Escoffier; Maïna Brus; Antoine D. Veron; Michel Khrestchatisky; François S. Roman

HIGHLIGHTSHippocampal lesions induced by the 4VO model can be predicted using anatomical MRI.The 4VO model induces strong neuronal death in the CA1 layer of dorsal hippocampus.BI ischemic rats develop amnesia with strong learning and memory impairments.Establishment of astroglial scars in CA1 layer from astrocytes and Nestin+ cells.Neurogenesis and cell proliferation enhanced in dentate gyrus after transient GCI. ABSTRACT Global Cerebral Ischemia (GCI) occurs following cardiac arrest or neonatal asphyxia and leads to harmful neurological consequences. In most cases, patients who survive cardiac arrest develop severe cognitive and motor impairments. This study focused on learning and memory deficits associated with brain neuroanatomical reorganization that appears after GCI. The four‐vessel occlusion (4VO) model was performed to produce a transient GCI. Hippocampal lesions in ischemic rats were visualized using anatomical Magnetic Resonance Imaging (aMRI). Then, the learning and memory abilities of control and ischemic (bilaterally or unilaterally) rats were assessed through the olfactory associated learning task. Finally, a “longitudinal” histological study was carried out to highlight the cellular reorganizations occurring after GCI. We demonstrated that the imaging, behavioral and histological results are closely related. In fact, aMRI revealed the appearance of hyper‐intense signals in the dorsal hippocampus at day 3 post‐GCI. Consequently, we showed a rise in cell proliferation (Ki 67+ cells) and endogenous neurogenesis especially in the dentate gyrus (DG) at day 3 post‐GCI. Then, hyper‐intense signals in the dorsal hippocampus were confirmed by strong neuronal losses in the CA1 layer at day 7 post‐GCI. These results were linked with severe learning and memory impairments only in bilaterally ischemic rats at day 14 post‐GCI. This amnesia was accompanied by huge astroglial and microglial hyperactivity at day 30 post‐GCI. Finally, Nestin+ cells and astrocytes gave rise to astroglial scars, which persisted 60 days post‐GCI. In the light of these results, the 4VO model appears a reliable method to produce amnesia in order to study and develop new therapeutic strategies.

Collaboration


Dive into the Stéphane D. Girard'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

Guy Escoffier

Aix-Marseille University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Santiago Rivera

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Anne Bernard

Aix-Marseille University

View shared research outputs
Top Co-Authors

Avatar

Arnaud Deveze

Aix-Marseille University

View shared research outputs
Researchain Logo
Decentralizing Knowledge