Network


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

Hotspot


Dive into the research topics where Corinne Marie is active.

Publication


Featured researches published by Corinne Marie.


Journal of Bacteriology | 2007

Control of EpsE, the Phosphoglycosyltransferase Initiating Exopolysaccharide Synthesis in Streptococcus thermophilus, by EpsD Tyrosine Kinase

Zoran Minic; Corinne Marie; Christine Delorme; Jean-Michel Faurie; Gérald Mercier; Dusko S. Ehrlich; Pierre Renault

We studied the roles of Streptococcus thermophilus phosphogalactosyltransferase (EpsE) (the priming enzyme), tyrosine kinase (EpsD), phosphatase (EpsB), and a membrane-associated protein with no known biochemical function (EpsC) in exopolysaccharide (EPS) synthesis. These proteins are well-conserved among bacteria and are usually encoded by clustered genes. Exopolysaccharide synthesis took place in the wild-type strain and a mutant lacking EpsB but not in mutants lacking EpsC, EpsD, or EpsE. The three mutants unable to synthesize EPS lacked the EpsE phosphogalactosyltransferase activity, while the two EPS-synthesizing strains possessed this activity, showing that EpsC and EpsD are required for EpsE function. An EpsD phosphorylated form was found in all strains except the epsC mutant, indicating that EpsC is necessary for EpsD phosphorylation. Moreover, the phosphorylated form of EpsD, a supposedly cytoplasmic protein, was found to be associated with the plasma membrane, possibly due to interaction with EpsC. Finally, the EpsD and EpsE elution profiles in a gel filtration chromatography assay were similar, suggesting that these two proteins colocalize in the membrane. Mutation of Tyr200, predicted to be a phosphorylation site and present in a conserved motif in bacterial phosphoglycosyltransferases, led to EpsE inactivation. In contrast, mutation of Tyr162 or Tyr199 had no effect. Taken together, these data show that EpsD controls EpsE activity. Possible mechanisms for this control are discussed.


Molecular Therapy | 2011

New generation of plasmid backbones devoid of antibiotic resistance marker for gene therapy trials.

Gaëlle Vandermeulen; Corinne Marie; Daniel Scherman; Véronique Préat

Since it has been established that the injection of plasmid DNA can lead to an efficient expression of a specific protein in vivo, nonviral gene therapy approaches have been considerably improved, allowing clinical trials. However, the use of antibiotic resistance genes as selection markers for plasmid production raises safety concerns which are often pointed out by the regulatory authorities. Indeed, a horizontal gene transfer to patients bacteria cannot be excluded, and residual antibiotic in the final product could provoke allergic reactions in sensitive individuals. A new generation of plasmid backbones devoid of antibiotic resistance marker has emerged to increase the safety profile of nonviral gene therapy trials. This article reviews the existing strategies for plasmid maintenance and, in particular, those that do not require the use of antibiotic resistance genes. They are based either on the complementation of auxotrophic strain, toxin-antitoxin systems, operator-repressor titration, RNA markers, or on the overexpression of a growth essential gene. Minicircles that allow removing of the antibiotic resistance gene from the initial vector will also be discussed. Furthermore, reported use of antibiotic-free plasmids in preclinical or clinical studies will be listed to provide a comprehensive view of these innovative technologies.


Journal of Gene Medicine | 2010

pFARs, Plasmids free of antibiotic resistance markers, display high-level transgene expression in muscle, skin and tumour cells

Corinne Marie; Gaëlle Vandermeulen; Mickael Quiviger; Magali Richard; Véronique Préat; Daniel Scherman

Nonviral gene therapy requires a high yield and a low cost production of eukaryotic expression vectors that meet defined criteria such as biosafety and quality of pharmaceutical grade. To fulfil these objectives, we designed a novel antibiotic‐free selection system.


Molecular therapy. Nucleic acids | 2017

Engineering of PEDF-Expressing Primary Pigment Epithelial Cells by the SB Transposon System Delivered by pFAR4 Plasmids

Gabriele Thumann; Nina Harmening; Cécile Prat-Souteyrand; Corinne Marie; Marie Pastor; Attila Sebe; Csaba Miskey; Laurence D. Hurst; Sabine Diarra; Martina Kropp; Peter Walter; Daniel Scherman; Zoltán Ivics; Zsuzsanna Izsvák; Sandra Johnen

Neovascular age-related macular degeneration (nvAMD) is characterized by choroidal blood vessels growing into the subretinal space, leading to retinal pigment epithelial (RPE) cell degeneration and vision loss. Vessel growth results from an imbalance of pro-angiogenic (e.g., vascular endothelial growth factor [VEGF]) and anti-angiogenic factors (e.g., pigment epithelium-derived factor [PEDF]). Current treatment using intravitreal injections of anti-VEGF antibodies improves vision in about 30% of patients but may be accompanied by side effects and non-compliance. To avoid the difficulties posed by frequent intravitreal injections, we have proposed the transplantation of pigment epithelial cells modified to overexpress human PEDF. Stable transgene integration and expression is ensured by the hyperactive Sleeping Beauty transposon system delivered by pFAR4 miniplasmids, which have a backbone free of antibiotic resistance markers. We demonstrated efficient expression of the PEDF gene and an optimized PEDF cDNA sequence in as few as 5 × 103 primary cells. At 3 weeks post-transfection, PEDF secretion was significantly elevated and long-term follow-up indicated a more stable secretion by cells transfected with the optimized PEDF transgene. Analysis of transgene insertion sites in human RPE cells showed an almost random genomic distribution. The results represent an important contribution toward a clinical trial aiming at a non-viral gene therapy of nvAMD.


Journal of Controlled Release | 2017

Cationic microbubbles and antibiotic-free miniplasmid for sustained ultrasound–mediated transgene expression in liver

Simona Manta; Gilles Renault; Anthony Delalande; Olivier Couture; Isabelle Lagoutte; Johanne Seguin; Franck Lager; Pascal Houzé; Patrick Midoux; Michel Bessodes; Daniel Scherman; Michel-Francis Bureau; Corinne Marie; Chantal Pichon; Nathalie Mignet

Despite the increasing number of clinical trials in gene therapy, no ideal methods still allow non-viral gene transfer in deep tissues such as the liver. We were interested in ultrasound (US)-mediated gene delivery to provide long term liver expression. For this purpose, new positively charged microbubbles were designed and complexed with pFAR4, a highly efficient small length miniplasmid DNA devoid of antibiotic resistance sequence. Sonoporation parameters, such as insonation time, acoustic pressure and duration of plasmid injection were controlled under ultrasound imaging guidance. The optimization of these various parameters was performed by bioluminescence optical imaging of luciferase reporter gene expression in the liver. Mice were injected with 50μg pFAR4-LUC either alone, or complexed with positively charged microbubbles, or co-injected with neutral MicroMarker™ microbubbles, followed by low ultrasound energy application to the liver. Injection of the pFAR4 encoding luciferase alone led to a transient transgene expression that lasted only for two days. The significant luciferase signal obtained with neutral microbubbles decreased over 2days and reached a plateau with a level around 1 log above the signal obtained with pFAR4 alone. With the newly designed positively charged microbubbles, we obtained a much stronger bioluminescence signal which increased over 2days. The 12-fold difference (p<0.05) between MicroMarker™ and our positively charged microbubbles was maintained over a period of 6months. Noteworthy, the positively charged microbubbles led to an improvement of 180-fold (p<0.001) as regard to free pDNA using unfocused ultrasound performed at clinically tolerated ultrasound amplitude. Transient liver damage was observed when using the cationic microbubble-pFAR4 complexes and the optimized sonoporation parameters. Immunohistochemistry analyses were performed to determine the nature of cells transfected. The pFAR4 miniplasmid complexed with cationic microbubbles allowed to transfect mostly hepatocytes compared to its co-injection with MicroMarker™ which transfected more preferentially endothelial cells.


Archive | 1994

Hrp and Avirulence Genes of Xanthomonas Campestris PV. Vesicatoria Controlling the Interaction with Pepper and Tomato

Ulla Bonas; Stefan Fenselau; Torsten Horns; Corinne Marie; Bernard Moussian; Michèle Pierre; Kai Wengelnik; Guido Van den Ackerveken

Xanthomonas campestris pathovar (pv.) vesicatoria (Xcv) is the causal agent of bacterial spot disease of pepper and tomato. The bacteria enter the plant tissue via stomata or wounds. After infection of a plant with Xcv two different types of reactions can be observed dependent on the genotype of both organisms. In the susceptible plant the infection with a virulent Xcv strain gives rise to watersoaked lesions that later on become necrotic (compatible interaction). Is the plant resistant and the bacterium avirulent, a hypersensitive response (HR) is induced (incompatible interaction). The HR is a local defense reaction accompanied by rapid necrosis of the infected tissue, leading to restriction of bacterial growth.


Molecular therapy. Nucleic acids | 2017

Long-Term PEDF Release in Rat Iris and Retinal Epithelial Cells after Sleeping Beauty Transposon-Mediated Gene Delivery

Laura Garcia-Garcia; Sergio Recalde; Maria Hernandez; Jaione Bezunartea; Juan R. Rodriguez-Madoz; Sandra Johnen; Sabine Diarra; Corinne Marie; Zsuzsanna Izsvák; Zoltán Ivics; Daniel Scherman; Martina Kropp; Gabriele Thumann; Felipe Prosper; Patricia Fernández-Robredo; Alfredo García-Layana

Pigment epithelium derived factor (PEDF) is a potent antiangiogenic, neurotrophic, and neuroprotective molecule that is the endogenous inhibitor of vascular endothelial growth factor (VEGF) in the retina. An ex vivo gene therapy approach based on transgenic overexpression of PEDF in the eye is assumed to rebalance the angiogenic-antiangiogenic milieu of the retina, resulting in growth regression of choroidal blood vessels, the hallmark of neovascular age-related macular degeneration. Here, we show that rat pigment epithelial cells can be efficiently transfected with the PEDF-expressing non-viral hyperactive Sleeping Beauty transposon system delivered in a form free of antibiotic resistance marker miniplasmids. The engineered retinal and iris pigment epithelium cells secrete high (141 ± 13 and 222 ± 14 ng) PEDF levels in 72 hr in vitro. In vivo studies showed cell survival and insert expression during at least 4 months. Transplantation of the engineered cells to the subretinal space of a rat model of choroidal neovascularization reduces almost 50% of the development of new vessels.


European Journal of Medical Genetics | 2018

Improved molecular platform for the gene therapy of rare diseases by liver protein secretion

Mickael Quiviger; Aristeidis Giannakopoulos; Sebastien Verhenne; Corinne Marie; Eleana F. Stavrou; Karen Vanhoorelbeke; Zsuzsanna Izsvák; Simon F. De Meyer; Aglaia Athanassiadou; Daniel Scherman

Many rare monogenic diseases are treated by protein replacement therapy, in which the missing protein is repetitively administered to the patient. However, in several cases, the missing protein is required at a high and sustained level, which renders protein therapy far from being adequate. As an alternative, a gene therapy treatment ensuring a sustained effectiveness would be particularly valuable. Liver is an optimal organ for the secretion and systemic distribution of a therapeutic transgene product. Cutting edge non-viral gene therapy tools were tested in order to produce a high and sustained level of therapeutic protein secretion by the liver using the hydrodynamic delivery technique. The use of S/MAR matrix attachment region provided a slight, however not statistically significant, increase in the expression of a reporter gene in the liver. We have selected the von Willebrand Factor (vWF) gene as a particularly challenging large gene (8.4 kb) for liver delivery and expression, and also because a high vWF blood concentration is required for disease correction. By using the optimized miniplasmid pFAR free of antibiotic resistance gene together with the Sleeping Beauty transposon and the hyperactive SB100X transposase, we have obtained a sustainable level of vWFblood secretion by the liver, at 65% of physiological level. Our results point to the general use of this plasmid platform using the liver as a protein factory to treat numerous rare disorders by gene therapy.


Molecular therapy. Nucleic acids | 2017

The Antibiotic-free pFAR4 Vector Paired with the Sleeping Beauty Transposon System Mediates Efficient Transgene Delivery in Human Cells

Marie Pastor; Sandra Johnen; Nina Harmening; Mickael Quiviger; Julie Pailloux; Martina Kropp; Peter Walter; Zoltán Ivics; Zsuzsanna Izsvák; Gabriele Thumann; Daniel Scherman; Corinne Marie

The anti-angiogenic and neurogenic pigment epithelium-derived factor (PEDF) demonstrated a potency to control choroidal neovascularization in age-related macular degeneration (AMD) patients. The goal of the present study was the development of an efficient and safe technique to integrate, ex vivo, the PEDF gene into retinal pigment epithelial (RPE) cells for later transplantation to the subretinal space of AMD patients to allow continuous PEDF secretion in the vicinity of the affected macula. Because successful gene therapy approaches require efficient gene delivery and stable gene expression, we used the antibiotic-free pFAR4 mini-plasmid vector to deliver the hyperactive Sleeping Beauty transposon system, which mediates transgene integration into the genome of host cells. In an initial study, lipofection-mediated co-transfection of HeLa cells with the SB100X transposase gene and a reporter marker delivered by pFAR4 showed a 2-fold higher level of genetically modified cells than when using the pT2 vectors. Similarly, with the pFAR4 constructs, electroporation-mediated transfection of primary human RPE cells led to 2.4-fold higher secretion of recombinant PEDF protein, which was still maintained 8 months after transfection. Thus, our results show that the pFAR4 plasmid is a superior vector for the delivery and integration of transgenes into eukaryotic cells.


Molecular Therapy | 2015

572. Antibiotic-Free Nonviral pFAR4 Vector Displays Efficient Transgene Delivery in Mouse and Human Cells

Marie Pastor; Sandra Johnen; Mickael Quiviger; Zsuzsanna Izsvák; Daniel Scherman; Gabriele Thumann; Corinne Marie

New generations of plasmid vectors used for non-viral gene therapy have a reduced size, thus allowing an increase in transfection efficiency and expression levels. We designed a novel vector, called pFAR4, which is Free of Antibiotic Resistance markers. In bacteria, the production of pFAR4 derivatives relies on a plasmid-borne function that suppresses a nonsense mutation introduced into an essential Escherichia coli gene.Thanks to its reduced size, pFAR4 appears to be an efficient gene vector as it displays a superior transgene expression level either after transfection of cultured B16 melanoma cells or electrotransfer into skin or tumors. Furthermore, hydrodynamic delivery into liver revealed that pFAR4 allowed a prolonged expression level, in comparison with kanamycin resistant plasmids that promote transgene silencing.We took advantage of the pFAR4 superiority in liver to assess a novel therapeutic approach for the Mucopolysaccharidosis type IIIA (MPS-IIIA) or Sanfilippo A syndrome. MPS-IIIA is a lysosomal storage genetic disease which results from the deficiency of the N-sulfoglucosamine sulfohydrolase (SGSH) protein, a sulfamidase required for the degradation of heparan sulfate glycosaminoglycans (GAGs). The accumulation of these macromolecules leads to somatic organ pathologies, severe neurodegeneration and patients’ death. In MPS-IIIA mice, hydrodynamic delivery of a pFAR4 derivative expressing the deficient enzyme from a liver-specific promoter allowed high and prolonged serum levels of sulfamidase protein that was efficiently taken-up by neighboring organs after its endocytosis by mannose 6-phosphate receptors-expressing cells. This led to the correction of GAG accumulation in all peripheral organs as well as in the brain, albeit at early stages of the disease. Improvement of our approach is provided by fusing the SGSH protein with peptides mediating transcytocis across the blood-brain barrier.Having established that pFAR4 is a potent expression vector in a non-integrative approach, our next step consisted in combining the pFAR4 technology and the Sleeping Beauty (SB) transposon system, which is composed of a transposase that excises the transgene-containing transposon from a donor plasmid and mediates its integration into the host cells’ genome. The reduced size of pFAR4 allowed an efficient delivery of SB components into human cells. Our current goal is to take the pFAR4 and Sleeping Beauty technologies to the clinic, in the context of an ex vivo gene therapy approach involving the transfection of autologous primary pigment epithelial cells for the treatment of neovascular age-related macular degeneration disease, in two phase I/II clinical trials.

Collaboration


Dive into the Corinne Marie's collaboration.

Top Co-Authors

Avatar

Daniel Scherman

Paris Descartes University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Zsuzsanna Izsvák

Max Delbrück Center for Molecular Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gaëlle Vandermeulen

Université catholique de Louvain

View shared research outputs
Top Co-Authors

Avatar

Véronique Préat

Université catholique de Louvain

View shared research outputs
Researchain Logo
Decentralizing Knowledge