Network


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

Hotspot


Dive into the research topics where Ariella Glasner is active.

Publication


Featured researches published by Ariella Glasner.


European Journal of Immunology | 2013

Mouse TIGIT inhibits NK-cell cytotoxicity upon interaction with PVR

Noa Stanietsky; Tihana Lenac Roviš; Ariella Glasner; Einat Seidel; Pinchas Tsukerman; Rachel Yamin; Jonatan Enk; Stipan Jonjić; Ofer Mandelboim

The activity of natural killer (NK) cells is controlled by a balance of signals derived from inhibitory and activating receptors. TIGIT is a novel inhibitory receptor, recently shown in humans to interact with two ligands: PVR and Nectin2 and to inhibit human NK‐cell cytotoxicity. Whether mouse TIGIT (mTIGIT) inhibits mouse NK‐cell cytotoxicity is unknown. Here we show that mTIGIT is expressed by mouse NK cells and interacts with mouse PVR. Using mouse and human Ig fusion proteins we show that while the human TIGIT (hTIGIT) cross‐reacts with mouse PVR (mPVR), the binding of mTIGIT is restricted to mPVR. We further demonstrate using surface plasmon resonance (SPR) and staining with Ig fusion proteins that mTIGIT binds to mPVR with higher affinity than the co‐stimulatory PVR‐binding receptor mouse DNAM1 (mDNAM1). Functionally, we show that triggering of mTIGIT leads to the inhibition of NK‐cell cytotoxicity, that IFN‐γ secretion is enhanced when mTIGIT is blocked and that the TIGIT‐mediated inhibition is dominant over the signals delivered by the PVR‐binding co‐stimulatory receptors. Additionally, we identify the inhibitory motif responsible for mTIGIT inhibition. In conclusion, we show that TIGIT is a powerful inhibitory receptor for mouse NK cells.


Journal of Immunology | 2012

Recognition and Prevention of Tumor Metastasis by the NK Receptor NKp46/NCR1

Ariella Glasner; Hormas Ghadially; Chamutal Gur; Noa Stanietsky; Pinchas Tsukerman; Jonatan Enk; Ofer Mandelboim

NK cells employ a variety of activating receptors to kill virally infected and tumor cells. Prominent among these receptors are the natural cytotoxicity receptors (NCRs) (NKp30, NKp44, and NKp46), of which only NKp46 has a mouse ortholog (NCR1). The tumor ligand(s) of NKp46/NCR1 is still unknown, but it was shown that the human NKp46 and the mouse NCR1 are involved in tumor eradication both in vitro and in vivo. Whether any of the NK activating receptors is involved in the prevention of tumor metastasis is unknown. To address this question, we studied the activity of the NK cell receptor NKp46/NCR1 in two spontaneous metastasis models, the B16F10.9 melanoma (B16) and the Lewis lung carcinoma (D122) in the NCR1 knockout mouse that was generated by our group, in various in vitro and in vivo assays. We demonstrated that all B16 and D122 tumors, including those generated in vivo, express an unknown ligand(s) for NKp46/NCR1. We have characterized the properties of the NKp46/NCR1 ligand(s) and demonstrated that NKp46/NCR1 is directly involved in the killing of B16 and D122 cells. Importantly, we showed in vivo that NKp46/NCR1 plays an important role in controlling B16 and D122 metastasis. Thus, to our knowledge, in this study we provide the first evidence for the direct involvement of a specific NK killer receptor in preventing tumor metastasis.


Journal of Virology | 2010

Killing of Avian and Swine Influenza Virus by Natural Killer Cells

Hagit Achdout; Tal Meningher; Shira Hirsh; Ariella Glasner; Yotam Bar-On; Chamutal Gur; Angel Porgador; Michal Mendelson; Michal Mandelboim; Ofer Mandelboim

ABSTRACT Today, global attention is focused on two influenza virus strains: the current pandemic strain, swine origin influenza virus (H1N1-2009), and the highly pathogenic avian influenza virus, H5N1. At present, the infection caused by the H1N1-2009 is moderate, with mortality rates of less <1%. In contrast, infection with the H5N1 virus resulted in high mortality rates, and ca. 60% of the infected patients succumb to the infection. Thus, one of the world greatest concerns is that the H5N1 virus will evolve to allow an efficient human infection and human-to-human transmission. Natural killer (NK) cells are one of the innate immune components playing an important role in fighting against influenza viruses. One of the major NK activating receptors involved in NK cell cytotoxicity is NKp46. We previously demonstrated that NKp46 recognizes the hemagglutinin proteins of B and A influenza virus strains. Whether NKp46 could also interact with H1N1-2009 virus or with the avian influenza virus is still unknown. We analyzed the immunological properties of both the avian and the H1N1-2009 influenza viruses. We show that NKp46 recognizes the hemagglutinins of H1N1-2009 and H5 and that this recognition leads to virus killing both in vitro and in vivo. However, importantly, while the swine H1-NKp46 interactions lead to the direct killing of the infected cells, the H5-NKp46 interactions were unable to elicit direct killing, probably because the NKp46 binding sites for these two viruses are different.


Cellular and Molecular Life Sciences | 2012

Virus-mediated inhibition of natural cytotoxicity receptor recognition

Einat Seidel; Ariella Glasner; Ofer Mandelboim

Natural killer (NK) cells are a part of the innate immune system that functions mainly to kill transformed and infected cells. Their activity is controlled by signals derived from a panel of activating and inhibitory receptors. The natural cytotoxicity receptors (NCRs): NKp30, NKp44, and NKp46 (NCR1 in mice) are prominent among the activating NK cell receptors and they are, notably, the only NK-activating receptors that are able to recognize pathogen-derived ligands. In addition, the NCRs also recognize cellular ligands, the identity of which remains largely unknown. In this review, we summarize the current knowledge regarding viruses that are recognized by the NCRs, focusing on the diverse immune-evasion mechanisms employed by viruses to escape this detection. We also discuss the unique role the NCRs have in regulating NK cell activity with particular emphasis on the in vivo function of NKp46/NCR1.


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.


PLOS ONE | 2012

Elucidating the Mechanisms of Influenza Virus Recognition by Ncr1

Ariella Glasner; Antonija Zurunic; Tal Meningher; Tihana Lenac Roviš; Pinchas Tsukerman; Yotam Bar-On; Rachel Yamin; Adrienne F.A. Meyers; Michal Mandeboim; Stipan Jonjić; Ofer Mandelboim

Natural killer (NK) cells are innate cytotoxic lymphocytes that specialize in the defense against viral infection and oncogenic transformation. Their action is tightly regulated by signals derived from inhibitory and activating receptors; the later include proteins such as the Natural Cytotoxicity Receptors (NCRs: NKp46, NKp44 and NKp30). Among the NCRs, NKp46 is the only receptor that has a mouse orthologue named Ncr1. NKp46/Ncr1 is also a unique marker expressed on NK and on Lymphoid tissue inducer (LTI) cells and it was implicated in the control of various viral infections, cancer and diabetes. We have previously shown that human NKp46 recognizes viral hemagglutinin (HA) in a sialic acid-dependent manner and that the O-glycosylation is essential for the NKp46 binding to viral HA. Here we studied the molecular interactions between Ncr1 and influenza viruses. We show that Ncr1 recognizes influenza virus in a sialic acid dependent manner and that N-glycosylation is important for this binding. Surprisingly we demonstrate that none of the predicted N-glycosilated residues of Ncr1 are essential for its binding to influenza virus and we thus conclude that other, yet unidentified N-glycosilated residues are responsible for its recognition. We have demonstrated that N glycosylation play little role in the recognition of mouse tumor cell lines and also showed the in-vivo importance of Ncr1 in the control of influenza virus infection by infecting C57BL/6 and BALB/c mice knockout for Ncr1 with influenza.


Cell Host & Microbe | 2013

Natural Killer Cell-Mediated Host Defense against Uropathogenic E. coli Is Counteracted by Bacterial HemolysinA-Dependent Killing of NK Cells

Chamutal Gur; Shunit Coppenhagen-Glazer; Shilo Rosenberg; Rachel Yamin; Jonatan Enk; Ariella Glasner; Yotam Bar-On; Omer Fleissig; Ronit Naor; Jawad Abed; Dror Mevorach; Zvi Granot; Gilad Bachrach; Ofer Mandelboim

Uropathogenic Escherichia coli (UPEC) are a common cause of urinary tract infections (UTIs) in humans. While the importance of natural killer (NK) cells in innate immune protection against tumors and viral infections is well documented, their role in defense against bacterial infections is still emerging, and their involvement in UPEC-mediated UTI is practically unknown. Using a systematic mutagenesis approach, we found that UPEC adheres to NK cells primarily via its type I fimbriae and employs its hemolysinA toxin to kill NK cells. In the absence of hemolysinA, NK cells directly respond to the bacteria and secrete the cytokine TNF-α, which results in decreased bacterial numbers in vitro and reduction of bacterial burden in the infected bladders. Thus, NK cells control UPEC via TNF-α production, which UPEC counteracts by hemolysinA-mediated killing of NK cells, representing a previously unrecognized host defense and microbial counterattack mechanism in the context of UTI.


European Journal of Immunology | 2013

NKp46 regulates allergic responses

Hormas Ghadially; Amjad Horani; Ariella Glasner; Moran Elboim; Roi Gazit; David Shoseyov; Ofer Mandelboim

Natural killer (NK) cells are cytotoxic cells that are able to rapidly kill viruses, tumor cells, parasites, bacteria, and even cells considered “self”. The activity of NK cells is controlled by a fine balance of inhibitory and activating signals mediated by a complex set of different receptors. However, the function of NK cells is not restricted only to the killing of target cells, NK cells also possess other properties such as the secretion of proangiogenic factors during pregnancy. Here, we demonstrate another unique NK‐cell activity, namely the regulation of T‐cell mediated allergic responses, which is dependent on the NK‐cell specific receptor NKp46 (Ncr1 in mice). Using mice in which the Ncr1 gene has been replaced with a green fluorescent protein, we demonstrate reduced delayed‐type hypersensitivity and airway hypersensitivity. Interestingly, we show that this reduction in airway hypersensitivity is due to differences in the stimulation of T cells resulting in an altered cytokine profile.


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.


Immunity | 2018

NKp46 Receptor-Mediated Interferon-γ Production by Natural Killer Cells Increases Fibronectin 1 to Alter Tumor Architecture and Control Metastasis

Ariella Glasner; Assi Levi; Jonatan Enk; Batya Isaacson; Sergey Viukov; Shari Orlanski; Alon Scope; Tzahi Neuman; Claes D. Enk; Jacob Hanna; Veronika Sexl; Stipan Jonjić; Barbara Seliger; Laurence Zitvogel; Ofer Mandelboim

Summary Natural killer (NK) cells are innate lymphoid cells, and their presence within human tumors correlates with better prognosis. However, the mechanisms by which NK cells control tumors in vivo are unclear. Here, we used reflectance confocal microscopy (RCM) imaging in humans and in mice to visualize tumor architecture in vivo. We demonstrated that signaling via the NK cell receptor NKp46 (human) and Ncr1 (mouse) induced interferon‐&ggr; (IFN‐&ggr;) secretion from intratumoral NK cells. NKp46‐ and Ncr1‐mediated IFN‐&ggr; production led to the increased expression of the extracellular matrix protein fibronectin 1 (FN1) in the tumors, which altered primary tumor architecture and resulted in decreased metastases formation. Injection of IFN‐&ggr; into tumor‐bearing mice or transgenic overexpression of Ncr1 in NK cells in mice resulted in decreased metastasis formation. Thus, we have defined a mechanism of NK cell‐mediated control of metastases in vivo that may help develop NK cell‐dependent cancer therapies. Graphical Abstract Figure. No Caption available. HighlightsNKp46 expression on NK cells controls growth of melanoma and carcinoma metastasesIn the absence of NKp46, tumor architectural properties indicate an aggressive phenotypeNK cell NKp46‐mediated IFN‐&ggr; production controls tumor structure via FN1 inductionIFN‐&ggr; treatment or Ncr1 overexpression in tumor‐bearing mice decreases tumor metastases &NA; NK cells defend against various pathogens and tumors, but the mechanisms by which they control tumor metastases are not clear. Here, Glasner et al. show that NK cells prevent tumor metastases in vivo by editing tumor architecture via NKp46‐mediated IFN‐&ggr; production that leads to upregulation of extracellular matrix protein FN1 in the tumor.

Collaboration


Dive into the Ariella Glasner's collaboration.

Top Co-Authors

Avatar

Ofer Mandelboim

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Yotam Bar-On

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Batya Isaacson

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

Rachel Yamin

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Pinchas Tsukerman

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Stipan Jonjić

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Alon Vitenshtein

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Jonatan Enk

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Orit Berhani

Hebrew University of Jerusalem

View shared research outputs
Researchain Logo
Decentralizing Knowledge