Network


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

Hotspot


Dive into the research topics where Olivier Disson is active.

Publication


Featured researches published by Olivier Disson.


PLOS Pathogens | 2008

A mouse model for Chikungunya: young age and inefficient type-I interferon signaling are risk factors for severe disease.

Thérèse Couderc; Fabrice Chrétien; Clémentine Schilte; Olivier Disson; Madly Brigitte; Florence Guivel-Benhassine; Yasmina Touret; Georges Barau; Nadège Cayet; Isabelle Schuffenecker; Philippe Desprès; Fernando Arenzana-Seisdedos; Alain Michault; Matthew L. Albert; Marc Lecuit

Chikungunya virus (CHIKV) is a re-emerging arbovirus responsible for a massive outbreak currently afflicting the Indian Ocean region and India. Infection from CHIKV typically induces a mild disease in humans, characterized by fever, myalgia, arthralgia, and rash. Cases of severe CHIKV infection involving the central nervous system (CNS) have recently been described in neonates as well as in adults with underlying conditions. The pathophysiology of CHIKV infection and the basis for disease severity are unknown. To address these critical issues, we have developed an animal model of CHIKV infection. We show here that whereas wild type (WT) adult mice are resistant to CHIKV infection, WT mouse neonates are susceptible and neonatal disease severity is age-dependent. Adult mice with a partially (IFN-α/βR+/−) or totally (IFN-α/βR−/−) abrogated type-I IFN pathway develop a mild or severe infection, respectively. In mice with a mild infection, after a burst of viral replication in the liver, CHIKV primarily targets muscle, joint, and skin fibroblasts, a cell and tissue tropism similar to that observed in biopsy samples of CHIKV-infected humans. In case of severe infections, CHIKV also disseminates to other tissues including the CNS, where it specifically targets the choroid plexuses and the leptomeninges. Together, these data indicate that CHIKV-associated symptoms match viral tissue and cell tropisms, and demonstrate that the fibroblast is a predominant target cell of CHIKV. These data also identify the neonatal phase and inefficient type-I IFN signaling as risk factors for severe CHIKV-associated disease. The development of a permissive small animal model will expedite the testing of future vaccines and therapeutic candidates.


Nature | 2008

Conjugated action of two species-specific invasion proteins for fetoplacental listeriosis.

Olivier Disson; Solène Grayo; Eugénie Huillet; Georgios Nikitas; Francina Langa-Vives; Olivier Dussurget; Marie Ragon; Alban Le Monnier; Charles Babinet; Pascale Cossart; Marc Lecuit

The ability to cross host barriers is an essential virulence determinant of invasive microbial pathogens. Listeria monocytogenes is a model microorganism that crosses human intestinal and placental barriers, and causes severe maternofetal infections by an unknown mechanism. Several studies have helped to characterize the bacterial invasion proteins InlA and InlB. However, their respective species specificity has complicated investigations on their in vivo role. Here we describe two novel and complementary animal models for human listeriosis: the gerbil, a natural host for L. monocytogenes, and a knock-in mouse line ubiquitously expressing humanized E-cadherin. Using these two models, we uncover the essential and interdependent roles of InlA and InlB in fetoplacental listeriosis, and thereby decipher the molecular mechanism underlying the ability of a microbe to target and cross the placental barrier.


Journal of Experimental Medicine | 2010

The surface protein HvgA mediates group B streptococcus hypervirulence and meningeal tropism in neonates

Asmaa Tazi; Olivier Disson; Samuel Bellais; Abdelouhab Bouaboud; Nicolas Dmytruk; Shaynoor Dramsi; Michel-Yves Mistou; Huot Khun; Charlotte Mechler; Isabelle Tardieux; Patrick Trieu-Cuot; Marc Lecuit; Claire Poyart

Lethal meningitis triggered by the hypervirulent group B streptococcus clone ST-17 is mediated by a novel surface protein called HvgA.


Immunity | 2015

Liver-Resident Macrophage Necroptosis Orchestrates Type 1 Microbicidal Inflammation and Type-2-Mediated Tissue Repair during Bacterial Infection

Camille Blériot; Théo Dupuis; Grégory Jouvion; Gérard Eberl; Olivier Disson; Marc Lecuit

Kupffer cells, the phagocytes of fetal origin that line the liver sinusoids, are key contributors of host defense against enteroinvasive bacteria. Here, we found that infection by Listeria monocytogenes induced the early necroptotic death of Kupffer cells, which was followed by monocyte recruitment and an anti-bacterial type 1 inflammatory response. Kupffer cell death also triggered a type 2 response that involved the hepatocyte-derived alarmin interleukin-33 (IL-33) and basophil-derived interleukin-4 (IL-4). This led to the alternative activation of the monocyte-derived macrophages recruited to the liver, which thereby replaced ablated Kupffer cells and restored liver homeostasis. Kupffer cell death is therefore a key signal orchestrating type 1 microbicidal inflammation and type-2-mediated liver repair upon infection. This indicates that beyond the classical dichotomy of type 1 and type 2 responses, these responses can develop sequentially in the context of a bacterial infection and act interdependently, orchestrating liver immune responses and return to homeostasis, respectively.


Emerging Infectious Diseases | 2010

Human Listeriosis Caused by Listeria ivanovii

Christelle Guillet; Olivier Join-Lambert; Alban Le Monnier; Alexandre Leclercq; Frédéric Méchaï; Marie-France Mamzer-Bruneel; Magdalena K. Bielecka; Mariela Scortti; Olivier Disson; Patrick Berche; José A. Vázquez-Boland; Olivier Lortholary; Marc Lecuit

Two species of Listeria are pathogenic; L. monocytogenes infects humans and animals, and L. ivanovii has been considered to infect ruminants only. We report L. ivanovii–associated gastroenteritis and bacteremia in a man. This isolate was indistinguishable from prototypic ruminant strains. L. ivanovii is thus an enteric opportunistic human pathogen.


Nature Genetics | 2016

Uncovering Listeria monocytogenes hypervirulence by harnessing its biodiversity

Mylène M. Maury; Yu-Huan Tsai; Caroline Charlier; Marie Touchon; Viviane Chenal-Francisque; Alexandre Leclercq; Alexis Criscuolo; Charlotte Gaultier; Sophie Roussel; Anne Brisabois; Olivier Disson; Eduardo P. C. Rocha; Sylvain Brisse; Marc Lecuit

Microbial pathogenesis studies are typically performed with reference strains, thereby overlooking within-species heterogeneity in microbial virulence. Here we integrated human epidemiological and clinical data with bacterial population genomics to harness the biodiversity of the model foodborne pathogen Listeria monocytogenes and decipher the basis of its neural and placental tropisms. Taking advantage of the clonal structure of this bacterial species, we identify clones epidemiologically associated either with food or with human central nervous system (CNS) or maternal-neonatal (MN) listeriosis. The latter clones are also most prevalent in patients without immunosuppressive comorbidities. Strikingly, CNS- and MN-associated clones are hypervirulent in a humanized mouse model of listeriosis. By integrating epidemiological data and comparative genomics, we have uncovered multiple new putative virulence factors and demonstrate experimentally the contribution of the first gene cluster mediating L. monocytogenes neural and placental tropisms. This study illustrates the exceptional power in harnessing microbial biodiversity to identify clinically relevant microbial virulence attributes.


Infection and Immunity | 2009

Real-Time Observation of Listeria monocytogenes-Phagocyte Interactions in Living Zebrafish Larvae†

Jean-Pierre Levraud; Olivier Disson; Karima Kissa; Isabelle Bonne; Pascale Cossart; Philippe Herbomel; Marc Lecuit

ABSTRACT The zebrafish, Danio rerio, has become a popular vertebrate model for the study of infections, mainly because of its excellent optical accessibility at the embryonic and larval stages, when the innate immune system is already effective. We have thus tested the susceptibility of zebrafish larvae to the human pathogen Listeria monocytogenes, a gram-positive, facultative, intracellular bacterium that is known to survive and multiply in professional phagocytes and that causes fatal meningitis and abortions. Intravenous injection of early zebrafish larvae resulted in a progressive and ultimately fatal infection. Blood-borne L. monocytogenes bacteria were quickly trapped and engulfed by macrophages, an event that, for the first time, could be captured in vivo and in real time. Granulocytes also participated in the innate immune response. As in mammals, bacteria could escape the macrophage phagosome in a listeriolysin-dependent manner and accessed the cytosol; this event was critical for bacterial virulence, as listeriolysin-deficient bacteria were completely avirulent. Actin comet tails and protrusions were observed, suggesting cell-to-cell spread; these phenomena also played a role in virulence in zebrafish larvae, as actA-deficient bacteria were attenuated. These results demonstrate the relevance of the genetically tractable and optically accessible zebrafish model for the study of L. monocytogenes pathogenesis and particularly for the dissection of its interactions with phagocytes in vivo, a key factor of L. monocytogenes virulence.


PLOS Pathogens | 2013

ActA Promotes Listeria monocytogenes Aggregation, Intestinal Colonization and Carriage

Laetitia Travier; Stéphanie Guadagnini; Edith Gouin; Alexandre Dufour; Viviane Chenal-Francisque; Pascale Cossart; Jean-Christophe Olivo-Marin; Jean-Marc Ghigo; Olivier Disson; Marc Lecuit

Listeria monocytogenes (Lm) is a ubiquitous bacterium able to survive and thrive within the environment and readily colonizes a wide range of substrates, often as a biofilm. It is also a facultative intracellular pathogen, which actively invades diverse hosts and induces listeriosis. So far, these two complementary facets of Lm biology have been studied independently. Here we demonstrate that the major Lm virulence determinant ActA, a PrfA-regulated gene product enabling actin polymerization and thereby promoting its intracellular motility and cell-to-cell spread, is critical for bacterial aggregation and biofilm formation. We show that ActA mediates Lm aggregation via direct ActA-ActA interactions and that the ActA C-terminal region, which is not involved in actin polymerization, is essential for aggregation in vitro. In mice permissive to orally-acquired listeriosis, ActA-mediated Lm aggregation is not observed in infected tissues but occurs in the gut lumen. Strikingly, ActA-dependent aggregating bacteria exhibit an increased ability to persist within the cecum and colon lumen of mice, and are shed in the feces three order of magnitude more efficiently and for twice as long than bacteria unable to aggregate. In conclusion, this study identifies a novel function for ActA and illustrates that in addition to contributing to its dissemination within the host, ActA plays a key role in Lm persistence within the host and in transmission from the host back to the environment.


Virulence | 2012

Targeting of the central nervous system by Listeria monocytogenes

Olivier Disson; Marc Lecuit

Among bacteria that reach the central nervous system (CNS), Listeria monocytogenes (Lm) is one of deadliest, in human and ruminant. This facultative intracellular bacterium has the particularity to induce meningitis, meningoencephalitis and rhombencephalitis. Mechanisms by which Lm accesses the CNS remain poorly understood, but two major routes of infection have been proposed, based on clinical, in vitro and in vivo observations. A retrograde neural route is likely to occur in ruminants upon crossing of the oral epithelium, and this probably accounts for the observation that Lm induces almost exclusively rhombencephalitis in these animals. In contrast, the hematogenous route is likely the most frequent in human, in whom bacteria circulating in the blood, either free or associated with leukocytes are thought to breach the blood-brain barrier. New animal models that faithfully reproduce the hallmarks of human neurolisterisosis will allow addressing the molecular mechanisms underlying Lm ability to induce CNS disease, and improve our understanding of the pathophysiology of this deadly infection.


Cold Spring Harbor Perspectives in Medicine | 2013

Concepts and Mechanisms: Crossing Host Barriers

Kelly S. Doran; Anirban Banerjee; Olivier Disson; Marc Lecuit

The human body is bordered by the skin and mucosa, which are the cellular barriers that define the frontier between the internal milieu and the external nonsterile environment. Additional cellular barriers, such as the placental and the blood-brain barriers, define protected niches within the host. In addition to their physiological roles, these host barriers provide both physical and immune defense against microbial infection. Yet, many pathogens have evolved elaborated mechanisms to target this line of defense, resulting in a microbial invasion of cells constitutive of host barriers, disruption of barrier integrity, and systemic dissemination and invasion of deeper tissues. Here we review representative examples of microbial interactions with human barriers, including the intestinal, placental, and blood-brain barriers, and discuss how these microbes adhere to, invade, breach, or compromise these barriers.

Collaboration


Dive into the Olivier Disson'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
Top Co-Authors

Avatar

Mylène M. Maury

Centre national de la recherche scientifique

View shared research outputs
Top Co-Authors

Avatar

Alain Michault

Necker-Enfants Malades Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge