Network


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

Hotspot


Dive into the research topics where Amélie Montel-Hagen is active.

Publication


Featured researches published by Amélie Montel-Hagen.


Cell | 2008

Erythrocyte Glut1 triggers dehydroascorbic acid uptake in mammals unable to synthesize vitamin C.

Amélie Montel-Hagen; Sandrina Kinet; Nicolas Manel; Cédric Mongellaz; Rainer Prohaska; Jean-Luc Battini; Jean Delaunay; Marc Sitbon; Naomi Taylor

Of all cells, human erythrocytes express the highest level of the Glut1 glucose transporter. However, the regulation and function of Glut1 during erythropoiesis are not known. Here, we report that glucose transport actually decreases during human erythropoiesis despite a >3-log increase in Glut1 transcripts. In contrast, Glut1-mediated transport of L-dehydroascorbic acid (DHA), an oxidized form of ascorbic acid (AA), is dramatically enhanced. We identified stomatin, an integral erythrocyte membrane protein, as regulating the switch from glucose to DHA transport. Notably though, we found that erythrocyte Glut1 and associated DHA uptake are unique traits of humans and the few other mammals that have lost the ability to synthesize AA from glucose. Accordingly, we show that mice, a species capable of synthesizing AA, express Glut4 but not Glut1 in mature erythrocytes. Thus, erythrocyte-specific coexpression of Glut1 with stomatin constitutes a compensatory mechanism in mammals that are unable to synthesize vitamin C.


Blood | 2008

The Glut1 and Glut4 glucose transporters are differentially expressed during perinatal and postnatal erythropoiesis

Amélie Montel-Hagen; Lionel Blanc; Myriam Boyer-Clavel; Chantal Jacquet; Michel Vidal; Marc Sitbon; Naomi Taylor

Glucose is a major source of energy for living organisms, and its transport in vertebrates is a universally conserved property. Of all cell lineages, human erythrocytes express the highest level of the Glut1 glucose transporter with more than 200,000 molecules per cell. However, we recently reported that erythrocyte Glut1 expression is a specific trait of vitamin C-deficient mammalian species, comprising only higher primates, guinea pigs, and fruit bats. Here, we show that in all other tested mammalian species, Glut1 was transiently expressed in erythrocytes during the neonatal period. Glut1 was up-regulated during the erythroblast stage of erythroid differentiation and was present on the vast majority of murine red blood cells (RBCs) at birth. Notably though, Glut1 was not induced in adult mice undergoing anemia-induced erythropoiesis, and under these conditions, the up-regulation of a distinct transporter, Glut4, was responsible for an increased glucose transport. Sp3 and Sp1 transcriptions factors have been proposed to regulate Glut1 transcription, and we find that the concomitant repression of Glut1 and induction of Glut4 was associated with a significantly augmented Sp3/Sp1 ratio. Glucose transporter expression patterns in mice and human erythrocytes are therefore distinct. In mice, there is a postnatal switch from Glut1 to Glut4, with Glut4 further up-regulated under anemic conditions.


Retrovirology | 2007

Isolated receptor binding domains of HTLV-1 and HTLV-2 envelopes bind Glut-1 on activated CD4+ and CD8+ T cells

Sandrina Kinet; Louise Swainson; Madakasira Lavanya; Cédric Mongellaz; Amélie Montel-Hagen; Marco Craveiro; Nicolas Manel; Jean-Luc Battini; Marc Sitbon; Naomi Taylor

BackgroundWe previously identified the glucose transporter Glut-1, a member of the multimembrane-spanning facilitative nutrient transporter family, as a receptor for both HTLV-1 and HTLV-2. However, a recent report concluded that Glut-1 cannot serve as a receptor for HTLV-1 on CD4 T cells: This was based mainly on their inability to detect Glut-1 on this lymphocyte subset using the commercial antibody mAb1418. It was therefore of significant interest to thoroughly assess Glut-1 expression on CD4 and CD8 T cells, and its association with HTLV-1 and -2 envelope binding.ResultsAs previously reported, ectopic expression of Glut-1 but not Glut-3 resulted in significantly augmented binding of tagged proteins harboring the receptor binding domains of either HTLV-1 or HTLV-2 envelope glycoproteins (H1RBD or H2RBD). Using antibodies raised against the carboxy-terminal peptide of Glut-1, we found that Glut-1 expression was significantly increased in both CD4 and CD8 cells following TCR stimulation. Corresponding increases in the binding of H1RBD as well as H2RBD, not detected on quiescent T cells, were observed following TCR engagement. Furthermore, increased Glut-1 expression was accompanied by a massive augmentation in glucose uptake in TCR-stimulated CD4 and CD8 lymphocytes. Finally, we determined that the apparent contradictory results obtained by Takenouchi et al were due to their monitoring of Glut-1 with a mAb that does not bind cells expressing endogenous Glut-1, including human erythrocytes that harbor 300,000 copies per cell.ConclusionTransfection of Glut-1 directly correlates with the capacities of HTLV-1 and HTLV-2 envelope-derived ligands to bind cells. Moreover, Glut-1 is induced by TCR engagement, resulting in massive increases in glucose uptake and binding of HTLV-1 and -2 envelopes to both CD4 and CD8 T lymphocytes. Therefore, Glut-1 is a primary binding receptor for HTLV-1 and HTLV-2 envelopes on activated CD4 as well as CD8 lymphocytes.


Current Opinion in Hematology | 2009

Erythroid glucose transporters.

Amélie Montel-Hagen; Marc Sitbon; Naomi Taylor

Purpose of reviewAnimals are heterotrophic and use sugar as their principal source of carbon. Every cell possesses at least one hexose transport system and of all cells, human erythrocytes express the highest level of the facilitative glucose transporter 1 (GLUT1). On the basis of human data, it was assumed that all mammalian erythrocytes express GLUT1 and that this transporter functions similarly in red cells of different species. Recent findingsAnalyses of erythrocytes from diverse mammalian species showed that GLUT1 is restricted to those few mammals who are unable to synthesize ascorbic acid from glucose comprising higher primates, guinea pigs, and fruit bats. In humans, erythroid differentiation results in a dramatic GLUT1-mediated increase in the transport of an oxidized form of vitamin C, L-dehydroascorbic acid. This preferential L-dehydroascorbic acid uptake is regulated by the association of GLUT1 with stomatin, an integral erythrocyte membrane protein. In species that produce ascorbic acid, erythroid GLUT1 expression appears to be limited to the fetal and neonatal period. In the case of murine erythrocytes, glucose transport function is thereafter achieved by GLUT4, a GLUT originally characterized by its sensitivity to insulin. SummaryRecent research has shown that erythrocyte expression of GLUT-type transporters varies between mammalian species and that their functions in this context can differ. These data identify new arrangements of GLUT members in red cell metabolism.


Journal of Immunology | 2008

Cell Surface Expression of the Bovine Leukemia Virus-Binding Receptor on B and T Lymphocytes Is Induced by Receptor Engagement

Madakasira Lavanya; Sandrina Kinet; Amélie Montel-Hagen; Cédric Mongellaz; Jean-Luc Battini; Marc Sitbon; Naomi Taylor

Bovine leukemia virus (BLV), one of the most common infectious viruses of cattle, is endemic in many herds. Approximately 30–40% of adult cows in the United States are infected by this oncogenic C-type retrovirus and 1–5% of animals will eventually develop a malignant lymphoma. BLV, like the human and simian T cell leukemia viruses, is a deltaretrovirus but, in contrast with the latter, the BLV receptor remains unidentified. In this study, we demonstrate that the amino-terminal 182 residues of the BLV envelope glycoprotein surface unit encompasses the receptor-binding domain. A bona fide interaction of this receptor-binding domain with the BLV receptor was demonstrated by specific interference with BLV, but not human T cell leukemia virus, envelope glycoprotein-mediated binding. We generated a rabbit Ig Fc-tagged BLV receptor-binding domain construct and ascertained that the ligand binds the BLV receptor on target cells from multiple species. Using this tool, we determined that the BLV-binding receptor is expressed on differentiating pro/pre-B cells in mouse bone marrow. However, the receptor was not detected on mature/quiescent B cells but was induced upon B cell activation. Activation of human B and T lymphocytes also induced surface BLV-binding receptor expression and required de novo protein synthesis. Receptor levels were down-regulated as activated lymphocytes returned to quiescence. In the human thymus, BLV-binding receptor expression was specifically detected on thymocytes responding to the IL-7 cytokine. Thus, expression of the BLV-binding receptor is a marker of enhanced metabolic activity in B cells, T cells, and thymocytes.


Cell | 2009

Response: Species Diversity in GLUT Expression and Function

Amélie Montel-Hagen; Sandrina Kinet; Nicolas Manel; Cédric Mongellaz; Rainer Prohaska; Jean-Luc Battini; Jean Delaunay; Marc Sitbon; Naomi Taylor

Of all cells in the human body, erythrocytes express the highest level of the GLUT1 glucose transporter, with more than 200,000 molecules per cell (Mueckler et al., 1985). We found that GLUT1 expression is significantly upregulated in late-stage erythroblasts, whereas glucose transport is decreased (Montel-Hagen et al., 2008a). This increase in GLUT1 expression was associated with enhanced transport of L-dehydroascorbic acid (DHA), an oxidized intermediate of ascorbic acid (AA). Moreover, the efficient capture of DHA by erythrocyte GLUT1 and its immediate reduction to AA constitutes a recycling system that has evolved in those mammals incapable of synthesizing vitamin C.


Methods of Molecular Biology | 2009

In Vivo and Ex Vivo Gene Transfer in Thymocytes and Thymocyte Precursors

Oumeya Adjali; Amélie Montel-Hagen; Louise Swainson; Sophie Marty; Rita Vicente; Cédric Mongellaz; Chantal Jacquet; Valérie S. Zimmermann; Naomi Taylor

The thymus provides a specialized environment allowing the differentiation of T lymphocytes from bone marrow-derived progenitor cells. We and others have demonstrated that gene transfer into distinct thymocyte populations can be obtained, both in vivo and ex vivo, using lentiviral vectors. Here, we describe techniques for intrathymic lentiviral transduction in mice, using a surgical approach wherein the thoracic cavity is exposed as well as a significantly less invasive strategy wherein virions are directly injected through the skin. Moreover, thymocyte differentiation from murine and human progenitors is now feasible in vitro, under conditions wherein the Notch and IL-7 signaling pathways are activated. We describe methods allowing transduction of murine and human progenitors and their subsequent differentiation into more mature thymocytes. Conditions for lentiviral gene transfer into more differentiated human thymocyte subsets are also presented. Optimization of technologies for HIV-based gene transfer into murine and human thymocyte progenitors will advance strategies aimed at modulating T-cell differentiation and function in-vivo; approaches potentially targeting patients with genetic and acquired immunodeficiencies as well as immune-sensitive tumors. Furthermore, this technology will foster the progression of basic research aimed at elucidating molecular aspects of T-cell differentiation in mice and humans.


Blood | 2008

Differential Glucose Transporter Expression during Perinatal and Postnatal Erythropoiesis

Amélie Montel-Hagen; Lionel Blanc; Myriam Boyer-Clavel; Chantal Jacquet; Michel Vidal; Marc Sitbon; Naomi Taylor


Archive | 2013

during perinatal and postnatal erythropoiesis The Glut1 and Glut4 glucose transporters are differentially expressed

Naomi Taylor; Amélie Montel-Hagen; Lionel Blanc; Myriam Boyer-Clavel; Chantal Jacquet; Michel Vidal; Marc Sitbon


Archive | 2011

transporter activity Overexpression of stomatin depresses GLUT-1 glucose

Wafa Abbud; Rainer Prohaska; Faramarz Ismail-Beigi; Guillaume Pilot; Dean F. Bushey; Réjane Pratelli; Glutamine Dumper; Marc Sitbon; Naomi Taylor; Amélie Montel-Hagen; Lionel Blanc; Myriam Boyer-Clavel; Chantal Jacquet; Michel Vidal; Mario Mairhofer; Marianne Steiner; Ulrich Salzer; Sidney M. Gospe; Sheila A. Baker; Vadim Y. Arshavsky

Collaboration


Dive into the Amélie Montel-Hagen's collaboration.

Top Co-Authors

Avatar

Marc Sitbon

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Naomi Taylor

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Cédric Mongellaz

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Chantal Jacquet

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Jean-Luc Battini

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Sandrina Kinet

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Myriam Boyer-Clavel

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Lionel Blanc

The Feinstein Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Louise Swainson

Centre national de la recherche scientifique

View shared research outputs
Researchain Logo
Decentralizing Knowledge