Network


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

Hotspot


Dive into the research topics where Yinon Levy is active.

Publication


Featured researches published by Yinon Levy.


PLOS ONE | 2009

Yersinia pestis Endowed with Increased Cytotoxicity Is Avirulent in a Bubonic Plague Model and Induces Rapid Protection against Pneumonic Plague

Ayelet Zauberman; Avital Tidhar; Yinon Levy; Erez Bar-Haim; Gideon Halperin; Yehuda Flashner; Sara Cohen; Avigdor Shafferman; Emanuelle Mamroud

An important virulence strategy evolved by bacterial pathogens to overcome host defenses is the modulation of host cell death. Previous observations have indicated that Yersinia pestis, the causative agent of plague disease, exhibits restricted capacity to induce cell death in macrophages due to ineffective translocation of the type III secretion effector YopJ, as opposed to the readily translocated YopP, the YopJ homologue of the enteropathogen Yersinia enterocolitica O∶8. This led us to suggest that reduced cytotoxic potency may allow pathogen propagation within a shielded niche, leading to increased virulence. To test the relationship between cytotoxic potential and virulence, we replaced Y. pestis YopJ with YopP. The YopP-expressing Y. pestis strain exhibited high cytotoxic activity against macrophages in vitro. Following subcutaneous infection, this strain had reduced ability to colonize internal organs, was unable to induce septicemia and exhibited at least a 107-fold reduction in virulence. Yet, upon intravenous or intranasal infection, it was still as virulent as the wild-type strain. The subcutaneous administration of the cytotoxic Y. pestis strain appears to activate a rapid and potent systemic, CTL-independent, immunoprotective response, allowing the organism to overcome simultaneous coinfection with 10,000 LD50 of virulent Y. pestis. Moreover, three days after subcutaneous administration of this strain, animals were also protected against septicemic or primary pneumonic plague. Our findings indicate that an inverse relationship exists between the cytotoxic potential of Y. pestis and its virulence following subcutaneous infection. This appears to be associated with the ability of the engineered cytotoxic Y. pestis strain to induce very rapid, effective and long-lasting protection against bubonic and pneumonic plague. These observations have novel implications for the development of vaccines/therapies against Y. pestis and shed new light on the virulence strategies of Y. pestis in nature.


Vaccine | 2008

Neutralization of Yersinia pestis-mediated macrophage cytotoxicity by anti-LcrV antibodies and its correlation with protective immunity in a mouse model of bubonic plague

Ayelet Zauberman; Sara Cohen; Yinon Levy; Gideon Halperin; Shirley Lazar; Baruch Velan; Avigdor Shafferman; Yehuda Flashner; Emanuelle Mamroud

Plague is a life-threatening disease caused by Yersinia pestis, for which effective-licensed vaccines and reliable predictors of in vivo immunity are lacking. V antigen (LcrV) is a major Y. pestis virulence factor that mediates translocation of the cytotoxic Yersinia protein effectors (Yops). It is a well-established protective antigen and a part of currently tested plague subunit vaccines. We have developed a highly sensitive in vitro macrophage cytotoxicity neutralization assay which is mediated by anti-LcrV antibodies; and studied the potential use of these neutralizing antibodies as an in vitro correlate of plague immunity in mice. The assay is based on a Y. pestis strain with enhanced cytotoxicity to macrophages in which endogenous yopJ was replaced by the more effectively translocated yopP of Y. enterocolitica O:8. Mice passively immunized with rabbit anti-LcrV IgG or actively immunized with recombinant LcrV were protected against lethal doses of a virulent Y. pestis strain, in a mouse model of bubonic plague. This protection significantly correlated with the in vitro neutralizing activity of the antisera but not with their corresponding ELISA titers. In actively immunized mice, a cutoff value for serum neutralizing activity, above which survival was assured with high degree of confidence, could be established for different vaccination regimes. The impact of overall findings on the potential use of serum neutralizing activity as a correlate of protective immunity is discussed.


PLOS Pathogens | 2015

Circumventing Y. pestis Virulence by Early Recruitment of Neutrophils to the Lungs during Pneumonic Plague

Yaron Vagima; Ayelet Zauberman; Yinon Levy; David Gur; Avital Tidhar; Moshe Aftalion; Avigdor Shafferman; Emanuelle Mamroud

Pneumonic plague is a fatal disease caused by Yersinia pestis that is associated with a delayed immune response in the lungs. Because neutrophils are the first immune cells recruited to sites of infection, we investigated the mechanisms responsible for their delayed homing to the lung. During the first 24 hr after pulmonary infection with a fully virulent Y. pestis strain, no significant changes were observed in the lungs in the levels of neutrophils infiltrate, expression of adhesion molecules, or the expression of the major neutrophil chemoattractants keratinocyte cell-derived chemokine (KC), macrophage inflammatory protein 2 (MIP-2) and granulocyte colony stimulating factor (G-CSF). In contrast, early induction of chemokines, rapid neutrophil infiltration and a reduced bacterial burden were observed in the lungs of mice infected with an avirulent Y. pestis strain. In vitro infection of lung-derived cell-lines with a YopJ mutant revealed the involvement of YopJ in the inhibition of chemoattractants expression. However, the recruitment of neutrophils to the lungs of mice infected with the mutant was still delayed and associated with rapid bacterial propagation and mortality. Interestingly, whereas KC, MIP-2 and G-CSF mRNA levels in the lungs were up-regulated early after infection with the mutant, their protein levels remained constant, suggesting that Y. pestis may employ additional mechanisms to suppress early chemoattractants induction in the lung. It therefore seems that prevention of the early influx of neutrophils to the lungs is of major importance for Y. pestis virulence. Indeed, pulmonary instillation of KC and MIP-2 to G-CSF-treated mice infected with Y. pestis led to rapid homing of neutrophils to the lung followed by a reduction in bacterial counts at 24 hr post-infection and improved survival rates. These observations shed new light on the virulence mechanisms of Y. pestis during pneumonic plague, and have implications for the development of novel therapies against this pathogen.


Frontiers in Cellular and Infection Microbiology | 2012

Early sensing of Yersinia pestis airway infection by bone marrow cells

Yaron Vagima; Yinon Levy; David Gur; Avital Tidhar; Moshe Aftalion; Hagar Abramovich; Eran Zahavy; Ayelet Zauberman; Yehuda Flashner; Avigdor Shafferman; Emanuelle Mamroud

Bacterial infection of the lungs triggers a swift innate immune response that involves the production of cytokines and chemokines that promote recruitment of immune cells from the bone marrow (BM) into the infected tissue and limit the ability of the pathogen to replicate. Recent in vivo studies of pneumonic plague in animal models indicate that the pulmonary pro-inflammatory response to airway infection with Yersinia pestis is substantially delayed in comparison to other pathogens. Consequently, uncontrolled proliferation of the pathogen in the lungs is observed, followed by dissemination to internal organs and death. While the lack of an adequate early immune response in the lung is well described, the response of BM-derived cells is poorly understood. In this study, we show that intranasal (i.n.) infection of mice with a fully virulent Y. pestis strain is sensed early by the BM compartment, resulting in a reduction in CXCR4 levels on BM neutrophils and their subsequent release into the blood 12 hours (h) post infection. In addition, increased levels of BM-derived hematopoietic stem and progenitor cells (HSPC) were detected in the blood early after infection. Mobilization of both immature and mature cells was accompanied by the reduction of BM SDF-1 (CXCL-12) levels and the reciprocal elevation of SDF-1 in the blood 24 h post infection. RT-PCR analysis of RNA collected from total BM cells revealed an early induction of myeloid-associated genes, suggesting a prompt commitment to myeloid lineage differentiation. These findings indicate that lung infection by Y. pestis is sensed by BM cells early after infection, although bacterial colonization of the BM occurs at late disease stages, and point on a potential cross-talk between the lung and the BM at early stages of pneumonic plague.


Microbial Cell Factories | 2006

Production and purification of high molecular weight oligomers of Yersinia pestis F1 capsular antigen released by high cell density culture of recombinant Escherichia coli cells carrying the caf1 operon

Tzvi Holtzman; Yinon Levy; Dino Marcus; Yehuda Flashner; Emanuelle Mamroud; Sara Cohen; Rephael Fass

Fermentation Expression of F1 by E. coli MC1060 (pBRK-F1) was studied in a 4-liter computer-controlled fermentor using a rich medium devoid of animal products: soy protein extract, trace elements and glycerol as carbon source (SY). This medium enabled efficient bacterial growth to a high cell density (60 OD600, Fig 1A). No loss of the expression plasmid was observed under these conditions. Unexpectedly, high and constant level of F1 expression was observed throughout the fermentation (up to 24 hours) even at 28°C (no temperature shift to 37°C was required, Fig. 1B). Moreover, at a high cell density (>10 OD600), F1 was released from the encapsulated E. coli cells and accumulated up to 0.9 g/L in the culture medium (Fig. 1C).


Frontiers in Cellular and Infection Microbiology | 2017

Host Iron Nutritional Immunity Induced by a Live Yersinia pestis Vaccine Strain Is Associated with Immediate Protection against Plague

Ayelet Zauberman; Yaron Vagima; Avital Tidhar; Moshe Aftalion; David Gur; Shahar Rotem; Theodor Chitlaru; Yinon Levy; Emanuelle Mamroud

Prompt and effective elicitation of protective immunity is highly relevant for cases of rapidly deteriorating fatal diseases, such as plague, which is caused by Yersinia pestis. Here, we assessed the potential of a live vaccine to induce rapid protection against this infection. We demonstrated that the Y. pestis EV76 live vaccine protected mice against an immediate lethal challenge, limiting the multiplication of the virulent pathogen and its dissemination into circulation. Ex vivo analysis of Y. pestis growth in serum derived from EV76-immunized mice revealed that an antibacterial activity was produced rapidly. This activity was mediated by the host heme- and iron-binding proteins hemopexin and transferrin, and it occurred in strong correlation with the kinetics of hemopexin induction in vivo. We suggest a new concept in which a live vaccine is capable of rapidly inducing iron nutritional immunity, thus limiting the propagation of pathogens. This concept could be exploited to design novel therapeutic interventions.


PLOS ONE | 2013

YopP-Expressing Variant of Y. pestis Activates a Potent Innate Immune Response Affording Cross-Protection against Yersiniosis and Tularemia

Ayelet Zauberman; Yehuda Flashner; Yinon Levy; Yaron Vagima; Avital Tidhar; Ofer Cohen; Erez Bar-Haim; David Gur; Moshe Aftalion; Gideon Halperin; Avigdor Shafferman; Emanuelle Mamroud

Plague, initiated by Yersinia pestis infection, is a rapidly progressing disease with a high mortality rate if not quickly treated. The existence of antibiotic-resistant Y. pestis strains emphasizes the need for the development of novel countermeasures against plague. We previously reported the generation of a recombinant Y. pestis strain (Kim53ΔJ+P) that over-expresses Y. enterocolitica YopP. When this strain was administered subcutaneously to mice, it elicited a fast and effective protective immune response in models of bubonic, pneumonic and septicemic plague. In the present study, we further characterized the immune response induced by the Kim53ΔJ+P recombinant strain. Using a panel of mouse strains defective in specific immune functions, we observed the induction of a prompt protective innate immune response that was interferon-γ dependent. Moreover, inoculation of mice with Y. pestis Kim53ΔJ+P elicited a rapid protective response against secondary infection by other bacterial pathogens, including the enteropathogen Y. enterocolitica and the respiratory pathogen Francisella tularensis. Thus, the development of new therapies to enhance the innate immune response may provide an initial critical delay in disease progression following the exposure to highly virulent bacterial pathogens, extending the time window for successful treatment.


The Journal of Infectious Diseases | 2016

Adjunctive Corticosteroid Treatment Against Yersinia pestis Improves Bacterial Clearance, Immunopathology and Survival in the Mouse Model of Bubonic Plague

Yinon Levy; Yaron Vagima; Avital Tidhar; Ayelet Zauberman; Moshe Aftalion; David Gur; Itay Fogel; Theodor Chitlaru; Yehuda Flashner; Emanuelle Mamroud

BACKGROUND Plague is initiated by Yersinia pestis, a highly virulent bacterial pathogen. In late stages of the infection, bacteria proliferate extensively in the internal organs despite the massive infiltration of neutrophils. The ineffective inflammatory response associated with tissue damage may contribute to the low efficacy of antiplague therapies during late stages of the infection. In the present study, we address the possibility of improving therapeutic efficacy by combining corticosteroid administration with antibody therapy in the mouse model of bubonic plague. METHODS Mice were subcutaneously infected with a fully virulent Y. pestis strain and treated at progressive stages of the disease with anti-Y. pestis antibodies alone or in combination with the corticosteroid methylprednisolone. RESULTS The addition of methylprednisolone to antibody therapy correlated with improved mouse survival, a significant decrease in the amount of neutrophils and matrix metalloproteinase 9 in the tissues, and the mitigation of tissue damage. Interestingly, the combined treatment led to a decrease in the bacterial loads in infected organs. CONCLUSIONS Corticosteroids induce an unexpectedly effective antibacterial response apart from their antiinflammatory properties, thereby improving treatment efficacy.


Archive | 2010

Protection Against Plague Afforded by Treatment with Polyclonal αLcrV and αF1 Antibodies

Yinon Levy; Yehuda Flashner; Ayelet Zauberman; Avital Tidhar; Moshe Aftalion; Shirley Lazar; David Gur; Sara Cohen; Avigdor Shafferman; Emanuelle Mamroud

Plague, initiated by Yersinia pestis infection, is a fatal disease that progresses rapidly and leads to high mortality rates if not treated within a short period of time after onset of symptoms. Antibiotics are effectively used for plague therapy yet antibiotic resistance Y. pestis strains have been reported and therefore alternative therapies are needed. In this study we demonstrate the ability of rabbit polyclonal antibodies directed against Y. pestis F1 and LcrV antigens to confer protection in mouse models of bubonic and pneumonic plague.


npj Vaccines | 2018

Targeting of the Yersinia pestis F1 capsular antigen by innate-like B1b cells mediates a rapid protective response against bubonic plague

Yinon Levy; Yaron Vagima; Avital Tidhar; Moshe Aftalion; David Gur; Uri Nili; Theodore Chitlaru; Ayelet Zauberman; Emanuelle Mamroud

The generation of adaptive immunity by vaccination is usually a prolonged process that requires multiple dosing over several months. Hence, vaccines are administered for disease prevention a relatively long time prior to possible infection as opposed to post-exposure prophylaxis, which typically requires rapid intervention such as antibiotic therapy. The emergence of pathogens resistant to common antibiotic treatments has prompted the search for alternative therapeutic strategies. We previously demonstrated that vaccination of mice with the F1 capsular antigen of Yersinia pestis elicits specific and effective yet, unexpectedly, rapid anti-plague immunity. Here, we show by applying genetic and immunological approaches that the F1 antigen is targeted by peritoneal innate-like B1b cells that generate a prompt T-independent (TI) anti-F1 humoral response. The rapid F1-mediated defense response was diminished in Xid (Btkm) mice in which B1 cell numbers and activity are limited. Binding of fluorophore-labeled F1 to peritoneal B1b cells was detected as soon as 6 h post vaccination, emphasizing the high speed of this process. By assessing the ability to achieve rapid immunity with monomerized F1, we show that the natural polymeric structure of F1 is essential for (i) rapid association with peritoneal B1b cells, (ii) early induction of anti-F1 titers and (iii) rapid TI immunity in the mouse model of bubonic plague. These observations shed new light on the potential of novel as well as well-known protective antigens in generating rapid immunity and could be implemented in the rational design of future vaccines.Bubonic plague: Mechanisms of a rapid antibody responseFull acquired immunity normally takes weeks to develop, however it’s possible to raise rapid (days) protective antibody responses against the bubonic plague bacterium Yersinia pestis by targeting its F1 capsular antigen. Emanuelle Mamroud and colleagues at the Institute of Biological Research in Israel use genetic and functional studies to understand the mechanism of this rapid anti-F1 antigen vaccine response. Subcutaneous vaccination with polymeric F1 antigen within hours results in selective binding to peritoneal B1b cells. Within seven days this generates an antibody response protective against fully virulent Y. pestis. This protection is spleen- and T-helper cell-independent but requires B1b cells. The form of F1 is also important, with monomerized antigen resulting in significantly inferior protection. These findings highlight the potential of post-exposure prophylaxis and the insights into its mechanism having wider implications for vaccine development.

Collaboration


Dive into the Yinon Levy's collaboration.

Top Co-Authors

Avatar

Emanuelle Mamroud

Israel Institute for Biological Research

View shared research outputs
Top Co-Authors

Avatar

Avital Tidhar

Israel Institute for Biological Research

View shared research outputs
Top Co-Authors

Avatar

Ayelet Zauberman

Israel Institute for Biological Research

View shared research outputs
Top Co-Authors

Avatar

David Gur

Israel Institute for Biological Research

View shared research outputs
Top Co-Authors

Avatar

Yehuda Flashner

Israel Institute for Biological Research

View shared research outputs
Top Co-Authors

Avatar

Avigdor Shafferman

Israel Institute for Biological Research

View shared research outputs
Top Co-Authors

Avatar

Moshe Aftalion

Israel Institute for Biological Research

View shared research outputs
Top Co-Authors

Avatar

Sara Cohen

Israel Institute for Biological Research

View shared research outputs
Top Co-Authors

Avatar

Yaron Vagima

Israel Institute for Biological Research

View shared research outputs
Top Co-Authors

Avatar

Gideon Halperin

Israel Institute for Biological Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge