Network


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

Hotspot


Dive into the research topics where Alon Vitenshtein is active.

Publication


Featured researches published by Alon Vitenshtein.


Cell Host & Microbe | 2011

An identical miRNA of the human JC and BK polyoma viruses targets the stress-induced ligand ULBP3 to escape immune elimination.

Yoav Bauman; Daphna Nachmani; Alon Vitenshtein; Pinchas Tsukerman; Nir Drayman; Noam Stern-Ginossar; Dikla Lankry; Raizy Gruda; Ofer Mandelboim

The human polyoma viruses JCV and BKV establish asymptomatic persistent infection in 65%-90% of humans but can cause severe illness under immunosuppressive conditions. The mechanisms by which these viruses evade immune recognition are unknown. Here we show that a viral miRNA identical in sequence between JCV and BKV targets the stress-induced ligand ULBP3, which is a protein recognized by the killer receptor NKG2D. Consequently, viral miRNA-mediated ULBP3 downregulation results in reduced NKG2D-mediated killing of virus-infected cells by natural killer (NK) cells. Importantly, when the activity of the viral miRNA was inhibited during infection, NK cells killed the infected cells more efficiently. Because NKG2D is also expressed by various T cell subsets, we propose that JCV and BKV use an identical miRNA that targets ULBP3 to escape detection by both the innate and adaptive immune systems, explaining how these viruses remain latent without being eliminated by the immune system.


Cell Reports | 2013

Neuraminidase-Mediated, NKp46-Dependent Immune-Evasion Mechanism of Influenza Viruses

Yotam Bar-On; Ariella Glasner; Tal Meningher; Hagit Achdout; Chamutal Gur; Dikla Lankry; Alon Vitenshtein; Adrienne F. A. Meyers; Michal Mandelboim; Ofer Mandelboim

Natural killer (NK) cells play an essential role in the defense against influenza virus, one of the deadliest respiratory viruses known today. The NKp46 receptor, expressed by NK cells, is critical for controlling influenza infections, as influenza-virus-infected cells are eliminated through the recognition of the viral hemagglutinin (HA) protein by NKp46. Here, we describe an immune-evasion mechanism of influenza viruses that is mediated by the neuraminidase (NA) protein. By using various NA blockers, we show that NA removes sialic acid residues from NKp46 and that this leads to reduced recognition of HA. Furthermore, we provide in vivo and in vitro evidence for the existence of this NA-mediated, NKp46-dependent immune-evasion mechanism and demonstrate that NA inhibitors, which are commonly used for the treatment of influenza infections, are useful not only as blockers of virus budding but also as boosters of NKp46 recognition.


Journal of Immunology | 2018

A Fusion Protein Encoding the Second Extracellular Domain of CCR5 Arrests Chemokine-Induced Cosignaling and Effectively Suppresses Ongoing Experimental Autoimmune Encephalomyelitis

Yair Sapir; Alon Vitenshtein; Yiftah Barsheshet; Yaniv Zohar; Gizi Wildbaum; Nathan Karin

CCR5 is a key CCR that is highly expressed on CD4+ T cells. It binds three different ligands: CCL3 (MIP-α), CCL4 (MIP-β), and CCL5 (RANTES). Recent studies suggested that the interaction between CCR5 and its ligands is essential not only for attracting these CCR5+ T cells but also substantial for transuding cosignals for their activation. The current study explores, for the first time, the in vivo consequences of CCR5 as a costimulatory molecule. First, we show redundancy between CCR5 ligands not only in chemoattractive properties but also in their ability to induced cosignals via CCR5. This has motivated us to generate a soluble receptor-based fusion protein that would selectively bind and neutralize all three CCR5 ligands. We show in this study that a 30-aa–based CCR5–Ig fusion protein encoding the second extracellular domain of receptor selectively binds and neutralizes all three CCR5 ligands and, when administered during ongoing experimental autoimmune encephalomyelitis, rapidly suppressed the disease while arresting Ag-specific effector T cell functions. Finally, our results clearly show that although CCR5 ligands induced cosignaling for IL-2 production is directed by CCR5, other proinflammatory properties of these ligands, such as TNF-α, IL-17, and IFN-γ production, are CCR5 independent and therefore likely to be mediated by the other receptors for these ligands. These findings imply that implementing a CCR5-Ig–based therapy would be advantageous over blockade of this receptor or of the use of mAbs for targeting a single CCR5 ligand.


PLOS Pathogens | 2013

The viral KSHV chemokine vMIP-II inhibits the migration of Naive and activated human NK cells by antagonizing two distinct chemokine receptors.

Rachel Yamin; Noa S. Kaynan; Ariella Glasner; Alon Vitenshtein; Pinchas Tsukerman; Yoav Bauman; Yael Ophir; Shlomo Elias; Yotam Bar-On; Chamutal Gur; Ofer Mandelboim

Natural killer (NK) cells are innate immune cells able to rapidly kill virus-infected and tumor cells. Two NK cell populations are found in the blood; the majority (90%) expresses the CD16 receptor and also express the CD56 protein in intermediate levels (CD56Dim CD16Pos) while the remaining 10% are CD16 negative and express CD56 in high levels (CD56Bright CD16Neg). NK cells also reside in some tissues and traffic to various infected organs through the usage of different chemokines and chemokine receptors. Kaposis sarcoma-associated herpesvirus (KSHV) is a human virus that has developed numerous sophisticated and versatile strategies to escape the attack of immune cells such as NK cells. Here, we investigate whether the KSHV derived cytokine (vIL-6) and chemokines (vMIP-I, vMIP-II, vMIP-III) affect NK cell activity. Using transwell migration assays, KSHV infected cells, as well as fusion and recombinant proteins, we show that out of the four cytokine/chemokines encoded by KSHV, vMIP-II is the only one that binds to the majority of NK cells, affecting their migration. We demonstrate that vMIP-II binds to two different receptors, CX3CR1 and CCR5, expressed by naïve CD56Dim CD16Pos NK cells and activated NK cells, respectively. Furthermore, we show that the binding of vMIP-II to CX3CR1 and CCR5 blocks the binding of the natural ligands of these receptors, Fractalkine (Fck) and RANTES, respectively. Finally, we show that vMIP-II inhibits the migration of naïve and activated NK cells towards Fck and RANTES. Thus, we present here a novel mechanism in which KSHV uses a unique protein that antagonizes the activity of two distinct chemokine receptors to inhibit the migration of naïve and activated NK cells.


Virology | 2015

Human cytomegalovirus induces a distinct innate immune response in the maternal-fetal interface.

Yiska Weisblum; Amos Panet; Zichria Zakay-Rones; Alon Vitenshtein; Ronit Haimov-Kochman; Debra Goldman-Wohl; Esther Oiknine-Djian; Rachel Yamin; Karen Meir; Hagai Amsalem; Tal Imbar; Ofer Mandelboim; Simcha Yagel; Dana G. Wolf

The initial interplay between human cytomegalovirus (HCMV) and innate tissue response in the human maternal-fetal interface, though crucial for determining the outcome of congenital HCMV infection, has remained unknown. We studied the innate response to HCMV within the milieu of the human decidua, the maternal aspect of the maternal-fetal interface, maintained ex vivo as an integral tissue. HCMV infection triggered a rapid and robust decidual-tissue innate immune response predominated by interferon (IFN)γ and IP-10 induction, dysregulating the decidual cytokine/chemokine environment in a distinctive fashion. The decidual-tissue response was already elicited during viral-tissue contact, and was not affected by neutralizing HCMV antibodies. Of note, IFNγ induction, reflecting immune-cell activation, was distinctive to the maternal decidua, and was not observed in concomitantly-infected placental (fetal) villi. Our studies in a clinically-relevant surrogate human model, provide a novel insight into the first-line decidual tissue response which could affect the outcome of congenital infection.


Cell Reports | 2016

CEACAM1-Mediated Inhibition of Virus Production

Alon Vitenshtein; Yiska Weisblum; Sebastian Hauka; Anne Halenius; Esther Oiknine-Djian; Pinchas Tsukerman; Yoav Bauman; Yotam Bar-On; Noam Stern-Ginossar; Jonatan Enk; Rona Ortenberg; Julie Tai; Gal Markel; Richard S. Blumberg; Hartmut Hengel; Stipan Jonjić; Dana G. Wolf; Heiko Adler; Robert Kammerer; Ofer Mandelboim

Summary Cells in our body can induce hundreds of antiviral genes following virus sensing, many of which remain largely uncharacterized. CEACAM1 has been previously shown to be induced by various innate systems; however, the reason for such tight integration to innate sensing systems was not apparent. Here, we show that CEACAM1 is induced following detection of HCMV and influenza viruses by their respective DNA and RNA innate sensors, IFI16 and RIG-I. This induction is mediated by IRF3, which bound to an ISRE element present in the human, but not mouse, CEACAM1 promoter. Furthermore, we demonstrate that, upon induction, CEACAM1 suppresses both HCMV and influenza viruses in an SHP2-dependent process and achieves this broad antiviral efficacy by suppressing mTOR-mediated protein biosynthesis. Finally, we show that CEACAM1 also inhibits viral spread in ex vivo human decidua organ culture.


Cell Reports | 2016

HCMV vCXCL1 Binds Several Chemokine Receptors and Preferentially Attracts Neutrophils over NK Cells by Interacting with CXCR2

Rachel Yamin; Laura S.M. Lecker; Yiska Weisblum; Alon Vitenshtein; Vu Thuy Khanh Le-Trilling; Dana G. Wolf; Ofer Mandelboim

HCMV is a highly sophisticated virus that has developed various mechanisms for immune evasion and viral dissemination throughout the body (partially mediated by neutrophils). NK cells play an important role in elimination of HCMV-infected cells. Both neutrophils and NK cells utilize similar sets of chemokine receptors to traffic, to and from, various organs. However, the mechanisms by which HCMV attracts neutrophils and not NK cells are largely unknown. Here, we show a unique viral protein, vCXCL1, which targets three chemokine receptors: CXCR1 and CXCR2 expressed on neutrophils and CXCR1 and CX3CR1 expressed on NK cells. Although vCXCL1 attracted both cell types, neutrophils migrated faster and more efficiently than NK cells through the binding of CXCR2. Therefore, we propose that HCMV has developed vCXCL1 to orchestrate its rapid systemic dissemination through preferential attraction of neutrophils and uses alternative mechanisms to counteract the later attraction of NK cells.


Oncotarget | 2016

Suppression of human metapneumovirus (HMPV) infection by the innate sensing gene CEACAM1

Mohammad Diab; Alon Vitenshtein; Yaron Drori; Rachel Yamin; Oded Danziger; Rachel Zamostiano; Michal Mandelboim; Eran Bacharach; Ofer Mandelboim

The innate sensing system is equipped with PRRs specialized in recognizing molecular structures (PAMPs) of various pathogens. This leads to the induction of anti-viral genes and inhibition of virus growth. Human Metapneumovirus (HMPV) is a major respiratory virus that causes an upper and lower respiratory tract infection in children. In this study we show that upon HMPV infection, the innate sensing system detects the viral RNA through the RIG-I sensor leading to induction of CEACAM1 expression. We further show that CEACAM1 is induced via binding of IRF3 to the CEACAM1 promoter. We demonstrate that induction of CEACAM1 suppresses the viral loads via inhibition of the translation machinery in the infected cells in an SHP2-dependent manner. In summary, we show here that HMPV-infected cells upregulates CEACAM1 to restrict HMPV infection.


Journal of Immunology | 2018

Decay of the Stress-Induced Ligand MICA Is Controlled by the Expression of an Alternative 3′ Untranslated Region

Tirtsah Toledano; Alon Vitenshtein; Noam Stern-Ginossar; Einat Seidel; Ofer Mandelboim

Recognition of the human stress-induced ligand MHC class I polypeptide-related sequence A (MICA) by the receptor NKG2D expressed on NK cells leads to NK cell–mediated killing of the target cells. Hence, the expression of MICA must be tightly regulated, and its cell surface expression needs to be quickly downregulated to avoid inappropriate activation of immune cells. In this article, we describe a transcript variant of human MICA that has not yet been studied, which contains a 3′ untranslated region of 119 nt instead of 174. We identify its polyadenylation signal and demonstrate that, upon stresses, such as heat shock, butyrate treatment, and some oxidative and DNA-damaging treatments, the balance between the two MICA variants changes in favor of the less stable, longer variant. Mechanistically, we showed that this change is linked to microRNA activity and that poly-ADP ribose polymerase 1 is involved in the induction of the longer variant following stress. Thus, to our knowledge, we identify the first regulatory mechanism of a stress ligand’s decay and also provide one of the first physiological examples for the biological function of a longer 3′ untranslated region of a particular gene.


Cancer Research | 2012

MiR-10b Downregulates the Stress-Induced Cell Surface Molecule MICB, a Critical Ligand for Cancer Cell Recognition by Natural Killer Cells

Pinchas Tsukerman; Noam Stern-Ginossar; Chamutal Gur; Ariella Glasner; Daphna Nachmani; Yoav Bauman; Rachel Yamin; Alon Vitenshtein; Noah Stanietsky; Tomer Bar-Mag; Dikla Lankry; Ofer Mandelboim

Collaboration


Dive into the Alon Vitenshtein's collaboration.

Top Co-Authors

Avatar

Ofer Mandelboim

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Rachel Yamin

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Yoav Bauman

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Ariella Glasner

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Chamutal Gur

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Noam Stern-Ginossar

Weizmann Institute of Science

View shared research outputs
Top Co-Authors

Avatar

Pinchas Tsukerman

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Dana G. Wolf

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Dikla Lankry

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Yiska Weisblum

Hebrew University of Jerusalem

View shared research outputs
Researchain Logo
Decentralizing Knowledge