Network


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

Hotspot


Dive into the research topics where Aurélie Chantôme is active.

Publication


Featured researches published by Aurélie Chantôme.


Molecular and Cellular Biology | 2003

HSP27 Is a Ubiquitin-Binding Protein Involved in I-κBα Proteasomal Degradation

Arnaud Parcellier; Elise Schmitt; Sandeep Gurbuxani; Daphné Seigneurin-Berny; Alena Pance; Aurélie Chantôme; Stéphanie Plenchette; Saadi Khochbin; Eric Solary; Carmen Garrido

ABSTRACT HSP27 is an ATP-independent chaperone that confers protection against apoptosis through various mechanisms, including a direct interaction with cytochrome c. Here we show that HSP27 overexpression in various cell types enhances the degradation of ubiquitinated proteins by the 26S proteasome in response to stressful stimuli, such as etoposide or tumor necrosis factor alpha (TNF-α). We demonstrate that HSP27 binds to polyubiquitin chains and to the 26S proteasome in vitro and in vivo. The ubiquitin-proteasome pathway is involved in the activation of transcription factor NF-κB by degrading its main inhibitor, I-κBα. HSP27 overexpression increases NF-κB nuclear relocalization, DNA binding, and transcriptional activity induced by etoposide, ΤNF-α, and interleukin 1β. HSP27 does not affect I-κBα phosphorylation but enhances the degradation of phosphorylated I-κBα by the proteasome. The interaction of HSP27 with the 26S proteasome is required to activate the proteasome and the degradation of phosphorylated I-κBα. A protein complex that includes HSP27, phosphorylated I-κBα, and the 26S proteasome is formed. Based on these observations, we propose that HSP27, under stress conditions, favors the degradation of ubiquitinated proteins, such as phosphorylated I-κBα. This novel function of HSP27 would account for its antiapoptotic properties through the enhancement of NF-κB activity.


Cancer Research | 2013

Pivotal role of the lipid raft SK3-Orai1 complex in human cancer cell migration and bone metastases

Aurélie Chantôme; Marie Potier-Cartereau; Lucie Clarysse; Gaëlle Fromont; Séverine Marionneau-Lambot; Maxime Guéguinou; Jean-Christophe Pagès; Christine Collin; Thibauld Oullier; Alban Girault; Flavie Arbion; Jean-Pierre Haelters; Michelle Pinault; Pierre Besson; Virginie Joulin; Philippe Bougnoux; Christophe Vandier

The SK3 channel, a potassium channel, was recently shown to control cancer cell migration, a critical step in metastasis outgrowth. Here, we report that expression of the SK3 channel was markedly associated with bone metastasis. The SK3 channel was shown to control constitutive Ca(2+) entry and cancer cell migration through an interaction with the Ca(2+) channel Orai1. We found that the SK3 channel triggers an association with the Orai1 channel within lipid rafts. This localization of an SK3-Orai1 complex seemed essential to control cancer cell migration. This suggests that the formation of this complex in lipid rafts is a gain-of-function, because we showed that none of the individual proteins were able to promote the complete phenotype. We identified the alkyl-lipid Ohmline as a disrupting agent for SK3-Orai1 lipid raft localization. Upon Ohmline treatment, the SK3-Orai1 complex moved away from lipid rafts, and SK3-dependent Ca(2+) entry, migration, and bone metastases were subsequently impaired. The colocalization of SK3 and Orai1 in primary human tumors and bone metastases further emphasized the clinical relevance of our observations. Targeting SK3-Orai1 in lipid rafts may inaugurate innovative approaches to inhibit bone metastases.


Experimental Cell Research | 2009

KCa2.3 channel-dependent hyperpolarization increases melanoma cell motility.

Aurélie Chantôme; Alban Girault; Marie Potier; Christine Collin; Pascal Vaudin; Jean-Christophe Pagès; Christophe Vandier; Virginie Joulin

Cell migration and invasion are required for tumour cells to spread from the primary tumour bed so as to form secondary tumours at distant sites. We report evidence of an unusual expression of KCa2.3 (SK3) protein in melanoma cell lines but not in normal melanocytes. Knockdown of the KCa2.3 channel led to plasma membrane depolarization, decreased 2D and 3D cell motility. Conversely, enforced production of KCa2.3 protein in KCa2.3 non-expressing cells led to the plasma membrane becoming hyperpolarized, and enhanced cell motility. In contrast, KCa3.1 channels had no effect on cell motility despite an active role in regulating membrane potential. Our data also suggest that membrane hyperpolarization increases melanoma cell motility and that this occurs through the KCa2.3 channel. Our findings reveal a previously unknown function of the KCa2.3 channel, and suggest that the KCa2.3 channel might be the only member of the Ca(2+)-activated K(+) channel family involved in melanoma cell motility pathways.


Biochimica et Biophysica Acta | 2014

KCa and Ca2 + channels: The complex thought ☆

Maxime Guéguinou; Aurélie Chantôme; Gaëlle Fromont; Philippe Bougnoux; Christophe Vandier; Marie Potier-Cartereau

Potassium channels belong to the largest and the most diverse super-families of ion channels. Among them, Ca(2+)-activated K(+) channels (KCa) comprise many members. Based on their single channel conductance they are divided into three subfamilies: big conductance (BKCa), intermediate conductance (IKCa) and small conductance (SKCa; SK1, SK2 and SK3). Ca(2+) channels are divided into two main families, voltage gated/voltage dependent Ca(2+) channels and non-voltage gated/voltage independent Ca(2+) channels. Based on their electrophysiological and pharmacological properties and on the tissue where there are expressed, voltage gated Ca(2+) channels (Cav) are divided into 5 families: T-type, L-type, N-type, P/Q-type and R-type Ca(2+). Non-voltage gated Ca(2+) channels comprise the TRP (TRPC, TRPV, TRPM, TRPA, TRPP, TRPML and TRPN) and Orai (Orai1 to Orai3) families and their partners STIM (STIM1 to STIM2). A depolarization is needed to activate voltage-gated Ca(2+) channels while non-voltage gated Ca(2+) channels are activated by Ca(2+) depletion of the endoplasmic reticulum stores (SOCs) or by receptors (ROCs). These two Ca(2+) channel families also control constitutive Ca(2+) entries. For reducing the energy consumption and for the fine regulation of Ca(2+), KCa and Ca(2+) channels appear associated as complexes in excitable and non-excitable cells. Interestingly, there is now evidence that KCa-Ca(2+) channel complexes are also found in cancer cells and contribute to cancer-associated functions such as cell proliferation, cell migration and the capacity to develop metastases. This article is part of a Special Issue entitled: Calcium signaling in health and disease. Guest Editors: Geert Bultynck, Jacques Haiech, Claus W. Heizmann, Joachim Krebs, and Marc Moreau.


Current Medicinal Chemistry | 2012

Targeting SKCa Channels in Cancer: Potential New Therapeutic Approaches

Alban Girault; Jean-Pierre Haelters; Marie Potier-Cartereau; Aurélie Chantôme; Philippe Bougnoux; Virginie Joulin; Christophe Vandier

Many studies have reported changes in potassium channel expression in many cancers and the involvement of these channels in various stages of cancer progression. By contrast, data concerning SKCa channels (small conductance calcium-activated potassium channels) have only recently become available. This review aims i) to present the structure and physiology of SKCa channels, ii) to provide an overview of published data concerning the SKCa proteins produced in tumor cells, and, whenever possible, the biological function assigned to them and iii) to review previous and novel modulators of SKCa channels. SKCa channels are activated by low concentrations of intracellular calcium and consist of homo- or heteromeric assemblies of α-subunits named SK1, SK2 and SK3. SK2-3 channels are expressed in tumors and have been assigned a biological function in cancer cells: the enhancement of cell proliferation and cell migration by hijacking the functions of SK2 and SK3 channels, respectively. Two major classes of SKCa modulators have been described: toxins (apamin) and small synthetic molecules. Most SKCa blockers are pore blockers, but some modify the calcium sensitivity of SKCa channels without interacting with the apamin binding site. In this review, we present edelfosine and ohmline as atypical anticancer agents and novel SK3 inhibitors. Edelfosine and ohmline are synthetic alkyl-lipids with structures different from all previously described SKCa modulators. They should pave the way for the development of a new class of migration-targeted anticancer agents. We believe that such blockers have potential for use in the prevention or treatment of metastasis.


British Journal of Pharmacology | 2011

The SK3/KCa2.3 potassium channel is a new cellular target for edelfosine

M. Potier; Aurélie Chantôme; Virginie Joulin; A. Girault; Sébastien Roger; Pierre Besson; M.-L. Jourdan; J.-Y. Leguennec; Philippe Bougnoux; Christophe Vandier

BACKGROUND AND PURPOSE The 1‐O‐octadecyl‐2‐O‐methyl‐sn‐glycero‐3‐phosphocholine (edelfosine) is an ether‐linked phospholipid with promising anti‐cancer properties but some side effects that preclude its full clinical therapeutic exploitation. We hypothesized that this lipid could interact with plasma membrane ion channels and modulate their function.


The FASEB Journal | 2000

Heat shock enhances transcriptional activation of the murine inducible nitric oxide synthase gene

Christopher E. Goldring; Sylvie Reveneau; Aurélie Chantôme; Alena Pance; Christophe Fleury; David A. Hume; David P. Sester; Bernard Mignotte; Jean-François Jeannin

There is considerable interest in determining the conditions leading to enhanced inducible nitric oxide synthase (iNOS) gene expression and nitric oxide (NO) biosynthesis. Using in vivofootprinting, we demonstrate that heat shock of murine macrophages concurrent with lipopolysaccharide (LPS) treatment stimulated changes in guanine methylation sensitivity at ‐898/9, at a putative partial heat shock element (HSE) and at ‐893/4, a site bordering an E‐box, within the iNOS gene enhancer, suggesting inducible occupation by transcription factors at these regions. LPS treatment accompanied by heat shock provoked increased iNOS gene transcription, increased levels of iNOS protein, and increased production of NO compared with LPS treatment alone. Electrophoretic mobility shift analysis revealed low constitutive levels of specific binding to an E‐box and a partial HSE within the iNOS enhancer. Binding to the E‐box was increased by LPS treatment or by heat shock, achieving a greater increase by a combination of both treatments. The proteins occupying this site were identified as belonging to the USF family of transcription factors. Heat shock or LPS increased binding to the HSE, and the factor responsible for this interaction was identified as heat shock factor‐1 (HSF‐1). Mutations at the HSE revealed the importance of HSF‐1 in the induction of iNOS by LPS. Thus, our data reveal two novel regulatory sites in the murine iNOS gene, one of which is implicated in enhancing iNOS expression via LPS stimulation, and provide the first evidence that heat shock enhances transcription of the iNOS gene. These results could have implications in the host response mechanism to fever‐associated gram‐negative infection.


The FASEB Journal | 2002

A repressor in the proximal human inducible nitric oxide synthase promoter modulates transcriptional activation

Alena Pance; Aurélie Chantôme; Sylvie Reveneau; Fatima Bentrari; Jean-François Jeannin

The human inducible nitric oxide synthase (iNOS or NOSII) gene is regulated through an extended and complex promoter. In this study, the transcriptional regulation of human NOSII is investigated in the human colon cell line HCT‐8R. Stimulation with a cytokine mix (interferon‐γ, interleukin 1‐β, and tumor necrosis factor α) induces NOSII mRNA accumulation, as well as promoter activity in these cells. Several random deletions were performed within the proximal 7 kb of the promoter, which led to the identification of a region, whose deletion provokes a marked increase in transcriptional activity upon cytokine stimulation. Furthermore, this region is shown to repress a viral‐driven luciferase construct, mainly at basal levels. An AP‐1‐like sequence present in this region that is specifically recognized by nuclear proteins is shown to be involved in the repressive effect. This element is capable of repressing a viral promoter, and its deletion augments cytokine‐stimulated transcription. These findings are confirmed in various cell lines and suggest a general mechanism for the control of basal levels of NOSII expression, to avoid unnecessary toxicity under normal conditions.


Biochimica et Biophysica Acta | 2015

Lipid rafts, KCa/ClCa/Ca2+ channel complexes and EGFR signaling: Novel targets to reduce tumor development by lipids?

Maxime Guéguinou; Audrey Gambade; Romain Felix; Aurélie Chantôme; Yann Fourbon; Philippe Bougnoux; Günther Weber; Marie Potier-Cartereau; Christophe Vandier

Membrane lipid rafts are distinct plasma membrane nanodomains that are enriched with cholesterol, sphingolipids and gangliosides, with occasional presence of saturated fatty acids and phospholipids containing saturated acyl chains. It is well known that they organize receptors (such as Epithelial Growth Factor Receptor), ion channels and their downstream acting molecules to regulate intracellular signaling pathways. Among them are Ca2+ signaling pathways, which are modified in tumor cells and inhibited upon membrane raft disruption. In addition to protein components, lipids from rafts also contribute to the organization and function of Ca2+ signaling microdomains. This article aims to focus on the lipid raft KCa/ClCa/Ca2+ channel complexes that regulate Ca2+ and EGFR signaling in cancer cells, and discusses the potential modification of these complexes by lipids as a novel therapeutic approach in tumor development. This article is part of a Special Issue entitled: Membrane channels and transporters in cancers.


Oncotarget | 2016

SK3/TRPC1/Orai1 complex regulates SOCE-dependent colon cancer cell migration: a novel opportunity to modulate anti-EGFR mAb action by the alkyl-lipid Ohmline

Maxime Guéguinou; Thomas Harnois; David Crottès; Arnaud Uguen; Nadine Déliot; Audrey Gambade; Aurélie Chantôme; Jean Pierre Haelters; Marie Lise Jourdan; Günther Weber; Olivier Soriani; Philippe Bougnoux; Olivier Mignen; Nicolas Bourmeyster; Bruno Constantin; Thierry Lecomte; Christophe Vandier; Marie Potier-Cartereau

Background Barely 10-20% of patients with metastatic colorectal cancer (mCRC) receive a clinical benefit from the use of anti-EGFR monoclonal antibodies (mAbs). We hypothesized that this could depends on their efficiency to reduce Store Operated Calcium Entry (SOCE) that are known to enhance cancer cells. Results In the present study, we demonstrate that SOCE promotes migration of colon cancer cell following the formation of a lipid raft ion channel complex composed of TRPC1/Orai1 and SK3 channels. Formation of this complex is stimulated by the phosphorylation of the reticular protein STIM1 by EGF and activation of the Akt pathway. Our data show that, in a positive feedback loop SOCE activates both Akt pathway and SK3 channel activity which lead to SOCE amplification. This amplification occurs through the activation of Rac1/Calpain mediated by Akt. We also show that Anti-EGFR mAbs can modulate SOCE and cancer cell migration through the Akt pathway. Interestingly, the alkyl-lipid Ohmline, which we previously showed to be an inhibitor of SK3 channel, can dissociated the lipid raft ion channel complex through decreased phosphorylation of Akt and modulation of mAbs action. Conclusions This study demonstrates that the inhibition of the SOCE-dependent colon cancer cell migration trough SK3/TRPC1/Orai1 channel complex by the alkyl-lipid Ohmline may be a novel strategy to modulate Anti-EGFR mAb action in mCRC.

Collaboration


Dive into the Aurélie Chantôme's collaboration.

Top Co-Authors

Avatar

Christophe Vandier

François Rabelais University

View shared research outputs
Top Co-Authors

Avatar

Philippe Bougnoux

François Rabelais University

View shared research outputs
Top Co-Authors

Avatar

Jean-François Jeannin

École pratique des hautes études

View shared research outputs
Top Co-Authors

Avatar

Sylvie Reveneau

École pratique des hautes études

View shared research outputs
Top Co-Authors

Avatar

Jean-Pierre Haelters

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Marie Potier-Cartereau

French Institute of Health and Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Charlotte M. Sevrain

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Hélène Couthon-Gourvès

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Alena Pance

Wellcome Trust Sanger Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge