Network


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

Hotspot


Dive into the research topics where Gérard Pierron is active.

Publication


Featured researches published by Gérard Pierron.


Molecular and Cellular Biology | 2005

Inhibition of Macroautophagy Triggers Apoptosis

Patricia Boya; Rosa-Ana Gonzalez-Polo; Noelia Casares; Jean-Luc Perfettini; Philippe Dessen; Nathanael Larochette; Didier Métivier; Daniel Meley; Sylvie Souquere; Tamotsu Yoshimori; Gérard Pierron; Patrice Codogno; Guido Kroemer

ABSTRACT Mammalian cells were observed to die under conditions in which nutrients were depleted and, simultaneously, macroautophagy was inhibited either genetically (by a small interfering RNA targeting Atg5, Atg6/Beclin 1-1, Atg10, or Atg12) or pharmacologically (by 3-methyladenine, hydroxychloroquine, bafilomycin A1, or monensin). Cell death occurred through apoptosis (type 1 cell death), since it was reduced by stabilization of mitochondrial membranes (with Bcl-2 or vMIA, a cytomegalovirus-derived gene) or by caspase inhibition. Under conditions in which the fusion between lysosomes and autophagosomes was inhibited, the formation of autophagic vacuoles was enhanced at a preapoptotic stage, as indicated by accumulation of LC3-II protein, ultrastructural studies, and an increase in the acidic vacuolar compartment. Cells exhibiting a morphology reminiscent of (autophagic) type 2 cell death, however, recovered, and only cells with a disrupted mitochondrial transmembrane potential were beyond the point of no return and inexorably died even under optimal culture conditions. All together, these data indicate that autophagy may be cytoprotective, at least under conditions of nutrient depletion, and point to an important cross talk between type 1 and type 2 cell death pathways.


The EMBO Journal | 2007

Functional and physical interaction between Bcl‐XL and a BH3‐like domain in Beclin‐1

M. Chiara Maiuri; Gaëtane Le Toumelin; Alfredo Criollo; Jean-Christophe Rain; Fabien Gautier; Philippe Juin; Ezgi Tasdemir; Gérard Pierron; Kostoula Troulinaki; Nektarios Tavernarakis; John Hickman; Olivier Geneste; Guido Kroemer

The anti‐apoptotic proteins Bcl‐2 and Bcl‐XL bind and inhibit Beclin‐1, an essential mediator of autophagy. Here, we demonstrate that this interaction involves a BH3 domain within Beclin‐1 (residues 114–123). The physical interaction between Beclin‐1 and Bcl‐XL is lost when the BH3 domain of Beclin‐1 or the BH3 receptor domain of Bcl‐XL is mutated. Mutation of the BH3 domain of Beclin‐1 or of the BH3 receptor domain of Bcl‐XL abolishes the Bcl‐XL‐mediated inhibition of autophagy triggered by Beclin‐1. The pharmacological BH3 mimetic ABT737 competitively inhibits the interaction between Beclin‐1 and Bcl‐2/Bcl‐XL, antagonizes autophagy inhibition by Bcl‐2/Bcl‐XL and hence stimulates autophagy. Knockout or knockdown of the BH3‐only protein Bad reduces starvation‐induced autophagy, whereas Bad overexpression induces autophagy in human cells. Gain‐of‐function mutation of the sole BH3‐only protein from Caenorhabditis elegans, EGL‐1, induces autophagy, while deletion of EGL‐1 compromises starvation‐induced autophagy. These results reveal a novel autophagy‐stimulatory function of BH3‐only proteins beyond their established role as apoptosis inducers. BH3‐only proteins and pharmacological BH3 mimetics induce autophagy by competitively disrupting the interaction between Beclin‐1 and Bcl‐2 or Bcl‐XL.


Nature Cell Biology | 2008

Regulation of autophagy by cytoplasmic p53

Ezgi Tasdemir; M. Chiara Maiuri; Lorenzo Galluzzi; Ilio Vitale; Mojgan Djavaheri-Mergny; Marcello D'Amelio; Alfredo Criollo; Eugenia Morselli; Changlian Zhu; Francis Harper; Ulf Nannmark; Chrysanthi Samara; Paolo Pinton; Jose Miguel Vicencio; Rosa Carnuccio; Ute M. Moll; Frank Madeo; Patrizia Paterlini-Bréchot; Rosario Rizzuto; Gérard Pierron; Klas Blomgren; Nektarios Tavernarakis; Patrice Codogno; Francesco Cecconi; Guido Kroemer

Multiple cellular stressors, including activation of the tumour suppressor p53, can stimulate autophagy. Here we show that deletion, depletion or inhibition of p53 can induce autophagy in human, mouse and nematode cells subjected to knockout, knockdown or pharmacological inhibition of p53. Enhanced autophagy improved the survival of p53-deficient cancer cells under conditions of hypoxia and nutrient depletion, allowing them to maintain high ATP levels. Inhibition of p53 led to autophagy in enucleated cells, and cytoplasmic, not nuclear, p53 was able to repress the enhanced autophagy of p53−/− cells. Many different inducers of autophagy (for example, starvation, rapamycin and toxins affecting the endoplasmic reticulum) stimulated proteasome-mediated degradation of p53 through a pathway relying on the E3 ubiquitin ligase HDM2. Inhibition of p53 degradation prevented the activation of autophagy in several cell lines, in response to several distinct stimuli. These results provide evidence of a key signalling pathway that links autophagy to the cancer-associated dysregulation of p53.


Journal of Experimental Medicine | 2005

Caspase-dependent immunogenicity of doxorubicin-induced tumor cell death

Noelia Casares; Marie O. Pequignot; Antoine Tesniere; François Ghiringhelli; Stephan Roux; Nathalie Chaput; Elise Schmitt; Ahmed Hamai; Sandra Hervas-Stubbs; Michel Obeid; Frédéric Coutant; Didier Métivier; Evelyne Pichard; Pierre Aucouturier; Gérard Pierron; Carmen Garrido; Laurence Zitvogel; Guido Kroemer

Systemic anticancer chemotherapy is immunosuppressive and mostly induces nonimmunogenic tumor cell death. Here, we show that even in the absence of any adjuvant, tumor cells dying in response to anthracyclins can elicit an effective antitumor immune response that suppresses the growth of inoculated tumors or leads to the regression of established neoplasia. Although both antracyclins and mitomycin C induced apoptosis with caspase activation, only anthracyclin-induced immunogenic cell death was immunogenic. Caspase inhibition by Z-VAD-fmk or transfection with the baculovirus inhibitor p35 did not inhibit doxorubicin (DX)-induced cell death, yet suppressed the immunogenicity of dying tumor cells in several rodent models of neoplasia. Depletion of dendritic cells (DCs) or CD8+T cells abolished the immune response against DX-treated apoptotic tumor cells in vivo. Caspase inhibition suppressed the capacity of DX-killed cells to be phagocytosed by DCs, yet had no effect on their capacity to elicit DC maturation. Freshly excised tumors became immunogenic upon DX treatment in vitro, and intratumoral inoculation of DX could trigger the regression of established tumors in immunocompetent mice. These results delineate a procedure for the generation of cancer vaccines and the stimulation of anti-neoplastic immune responses in vivo.


Journal of Cell Science | 2005

The apoptosis/autophagy paradox: autophagic vacuolization before apoptotic death

Rosa-Ana Gonzalez-Polo; Patricia Boya; Anne-Laure Pauleau; Abdelali Jalil; Nathanael Larochette; Sylvie Souquere; Eeva-Liisa Eskelinen; Gérard Pierron; Paul Saftig; Guido Kroemer

Autophagic cell death is morphologically characterized by an accumulation of autophagic vacuoles. Here, we show that inactivation of LAMP2 by RNA interference or by homologous recombination leads to autophagic vacuolization in nutrient-depleted cells. Cells that lack LAMP2 expression showed an enhanced accumulation of vacuoles carrying the marker LC3, yet a decreased colocalization of LC3 and lysosomes, suggesting that the fusion between autophagic vacuoles and lysosomes was inhibited. While a fraction of mitochondria from starved LAMP2-expressing cells colocalized with lysosomal markers, within autophagolysosomes, no such colocalization was found on removal of LAMP2 from the experimental system. Of note, LAMP1 depletion had no such effects and did not aggravate the phenotype induced by LAMP2-specific small interfering RNA. Serum and amino acid-starved LAMP2-negative cells exhibited an accumulation of autophagic vacuoles and then succumbed to cell death with hallmarks of apoptosis such as loss of the mitochondrial transmembrane potential, caspase activation and chromatin condensation. While caspase inhibition retarded cell death, it had no protective effect on mitochondria. Stabilization of mitochondria by overexpression of Bcl-2 or the mitochondrion-targeted cytomegalovirus protein vMIA, however, blocked all signs of apoptosis. Neither caspase inhibition nor mitochondrial stabilization antagonized autophagic vacuolization in LAMP2-deficient cells. Altogether, these data indicate that accumulation of autophagic vacuoles can precede apoptotic cell death. These findings argue against the clear-cut distinction between type 1 (apoptotic) and type 2 (autophagic) cell death.


The EMBO Journal | 2009

Mechanisms of pre‐apoptotic calreticulin exposure in immunogenic cell death

Theocharis Panaretakis; Oliver Kepp; Ulf Brockmeier; Antoine Tesniere; Ann-Charlotte Björklund; Daniel C. Chapman; Michael Durchschlag; Nicholas Joza; Gérard Pierron; Peter van Endert; Junying Yuan; Laurence Zitvogel; Frank Madeo; David B. Williams; Guido Kroemer

Dying tumour cells can elicit a potent anticancer immune response by exposing the calreticulin (CRT)/ERp57 complex on the cell surface before the cells manifest any signs of apoptosis. Here, we enumerate elements of the pathway that mediates pre‐apoptotic CRT/ERp57 exposure in response to several immunogenic anticancer agents. Early activation of the endoplasmic reticulum (ER)‐sessile kinase PERK leads to phosphorylation of the translation initiation factor eIF2α, followed by partial activation of caspase‐8 (but not caspase‐3), caspase‐8‐mediated cleavage of the ER protein BAP31 and conformational activation of Bax and Bak. Finally, a pool of CRT that has transited the Golgi apparatus is secreted by SNARE‐dependent exocytosis. Knock‐in mutation of eIF2α (to make it non‐phosphorylatable) or BAP31 (to render it uncleavable), depletion of PERK, caspase‐8, BAP31, Bax, Bak or SNAREs abolished CRT/ERp57 exposure induced by anthracyclines, oxaliplatin and ultraviolet C light. Depletion of PERK, caspase‐8 or SNAREs had no effect on cell death induced by anthracyclines, yet abolished the immunogenicity of cell death, which could be restored by absorbing recombinant CRT to the cell surface.


Journal of Biological Chemistry | 2006

NF-kappa B activation represses TNF alpha-induced autophagy

Mojgan Djavaheri-Mergny; Manuela Amelotti; Julie Mathieu; Françoise Besançon; Chantal Bauvy; Sylvie Souquere; Gérard Pierron; Patrice Codogno

Activation of NF-κB and autophagy are two processes involved in the regulation of cell death, but the possible cross-talk between these two signaling pathways is largely unknown. Here, we show that NF-κB activation mediates repression of autophagy in tumor necrosis factor-α (TNFα)-treated Ewing sarcoma cells. This repression is associated with an NF-κB-dependent activation of the autophagy inhibitor mTOR. In contrast, in cells lacking NF-κB activation, TNFα treatment up-regulates the expression of the autophagy-promoting protein Beclin 1 and subsequently induces the accumulation of autophagic vacuoles. Both of these responses are dependent on reactive oxygen species (ROS) production and can be mimicked in NF-κB-competent cells by the addition of H2O2. Small interfering RNA-mediated knockdown of beclin 1 and atg7 expression, two autophagy-related genes, reduced TNFα- and reactive oxygen species-induced apoptosis in cells lacking NF-κB activation and in NF-κB-competent cells, respectively. These findings demonstrate that autophagy may amplify apoptosis when associated with a death signaling pathway. They are also evidence that inhibition of autophagy is a novel mechanism of the antiapoptotic function of NF-κB activation. We suggest that stimulation of autophagy may be a potential way bypassing the resistance of cancer cells to anti-cancer agents that activate NF-κB.


Cell Death & Differentiation | 2007

Regulation of autophagy by the inositol trisphosphate receptor.

Alfredo Criollo; Maria Chiara Maiuri; Ezgi Tasdemir; I Vitale; A. A. Fiebig; David W. Andrews; Jordi Molgó; Sergio Lavandero; Francis Harper; Gérard Pierron; D. Di Stefano; Rosario Rizzuto; Guido Kroemer

The reduction of intracellular 1,4,5-inositol trisphosphate (IP3) levels stimulates autophagy, whereas the enhancement of IP3 levels inhibits autophagy induced by nutrient depletion. Here, we show that knockdown of the IP3 receptor (IP3R) with small interfering RNAs and pharmacological IP3R blockade is a strong stimulus for the induction of autophagy. The IP3R is known to reside in the membranes of the endoplasmic reticulum (ER) as well as within ER–mitochondrial contact sites, and IP3R blockade triggered the autophagy of both ER and mitochondria, as exactly observed in starvation-induced autophagy. ER stressors such as tunicamycin and thapsigargin also induced autophagy of ER and, to less extent, of mitochondria. Autophagy triggered by starvation or IP3R blockade was inhibited by Bcl-2 and Bcl-XL specifically targeted to ER but not Bcl-2 or Bcl-XL proteins targeted to mitochondria. In contrast, ER stress-induced autophagy was not inhibited by Bcl-2 and Bcl-XL. Autophagy promoted by IP3R inhibition could not be attributed to a modulation of steady-state Ca2+ levels in the ER or in the cytosol, yet involved the obligate contribution of Beclin-1, autophagy-related gene (Atg)5, Atg10, Atg12 and hVps34. Altogether, these results strongly suggest that IP3R exerts a major role in the physiological control of autophagy.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Syncytin-A knockout mice demonstrate the critical role in placentation of a fusogenic, endogenous retrovirus-derived, envelope gene

Anne Dupressoir; Cécile Vernochet; Olivia Bawa; Francis Harper; Gérard Pierron; Paule Opolon; Thierry Heidmann

In most mammalian species, a key process of placenta development is the fusion of trophoblast cells into a highly specialized, multinucleated syncytiotrophoblast layer, through which most of the maternofetal exchanges take place. Little is known about this process, despite the recent identification of 2 pairs of envelope genes of retroviral origin, independently acquired by the human (syncytin-1 and syncytin-2) and mouse (syncytin-A and syncytin-B) genomes, specifically expressed in the placenta, and with in vitro cell–cell fusion activity. By generating knockout mice, we show here that homozygous syncytin-A null mouse embryos die in utero between 11.5 and 13.5 days of gestation. Refined cellular and subcellular analyses of the syncytin-A-deficient placentae disclose specific disruption of the architecture of the syncytiotrophoblast-containing labyrinth, with the trophoblast cells failing to fuse into an interhemal syncytial layer. Lack of syncytin-A-mediated trophoblast cell fusion is associated with cell overexpansion at the expense of fetal blood vessel spaces and with apoptosis, adding to the observed maternofetal interface structural defects to provoke decreased vascularization, inhibition of placental transport, and fetal growth retardation, ultimately resulting in death of the embryo. These results demonstrate that syncytin-A is essential for trophoblast cell differentiation and syncytiotrophoblast morphogenesis during placenta development, and they provide evidence that genes captured from ancestral retroviruses have been pivotal in the acquisition of new, important functions in mammalian evolution.


The EMBO Journal | 2010

The IKK complex contributes to the induction of autophagy

Alfredo Criollo; Laura Senovilla; Hélène Authier; Maria Chiara Maiuri; Eugenia Morselli; Ilio Vitale; Oliver Kepp; Ezgi Tasdemir; Lorenzo Galluzzi; Shensi Shen; Nicolas F. Delahaye; Antoine Tesniere; Daniela De Stefano; Amena Ben Younes; Francis Harper; Gérard Pierron; Sergio Lavandero; Laurence Zitvogel; Alain Israël; Véronique Baud; Guido Kroemer

In response to stress, cells start transcriptional and transcription‐independent programs that can lead to adaptation or death. Here, we show that multiple inducers of autophagy, including nutrient depletion, trigger the activation of the IKK (IκB kinase) complex that is best known for its essential role in the activation of the transcription factor NF‐κB by stress. Constitutively active IKK subunits stimulated autophagy and transduced multiple signals that operate in starvation‐induced autophagy, including the phosphorylation of AMPK and JNK1. Genetic inhibition of the nuclear translocation of NF‐κB or ablation of the p65/RelA NF‐κB subunit failed to suppress IKK‐induced autophagy, indicating that IKK can promote the autophagic pathway in an NF‐κB‐independent manner. In murine and human cells, knockout and/or knockdown of IKK subunits (but not that of p65) prevented the induction of autophagy in response to multiple stimuli. Moreover, the knockout of IKK‐β suppressed the activation of autophagy by food deprivation or rapamycin injections in vivo, in mice. Altogether, these results indicate that IKK has a cardinal role in the stimulation of autophagy by physiological and pharmacological stimuli.

Collaboration


Dive into the Gérard Pierron's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Francis Harper

Centre national de la recherche scientifique

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
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ilio Vitale

University of Rome Tor Vergata

View shared research outputs
Researchain Logo
Decentralizing Knowledge