Network


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

Hotspot


Dive into the research topics where Benyamin Rosental is active.

Publication


Featured researches published by Benyamin Rosental.


Journal of Immunology | 2009

NKp44 Receptor Mediates Interaction of the Envelope Glycoproteins from the West Nile and Dengue Viruses with NK Cells

Oren Hershkovitz; Benyamin Rosental; Lior Rosenberg; Martha Erika Navarro-Sanchez; Sergey Jivov; Alon Zilka; Orly Gershoni-Yahalom; Elodie Brient-Litzler; Hugues Bedouelle; Joanna W. Ho; Kerry S. Campbell; Bracha Rager-Zisman; Philippe Desprès; Angel Porgador

Dengue virus (DV) and West Nile virus (WNV) have become a global concern due to their widespread distribution and their ability to cause a variety of human diseases. Antiviral immune defenses involve NK cells. In the present study, we investigated the interaction between NK cells and these two flaviviruses. We show that the NK-activating receptor NKp44 is involved in virally mediated NK activation through direct interaction with the flavivirus envelope protein. Recombinant NKp44 directly binds to purified DV and WNV envelope proteins and specifically to domain III of WNV envelope protein; it also binds to WNV virus-like particles. These WNV-virus-like particles and WNV-domain III of WNV envelope protein directly bind NK cells expressing high levels of NKp44. Functionally, interaction of NK cells with infective and inactivated WNV results in NKp44-mediated NK degranulation. Finally, WNV infection of cells results in increased binding of rNKp44 that is specifically inhibited by anti-WNV serum. WNV-infected target cells induce IFN-γ secretion and augmented lysis by NKp44-expressing primary NK cells that are blocked by anti-NKp44 Abs. Our findings show that triggering of NK cells by flavivirus is mediated by interaction of NKp44 with the flavivirus envelope protein.


Journal of Immunology | 2011

Proliferating Cell Nuclear Antigen Is a Novel Inhibitory Ligand for the Natural Cytotoxicity Receptor NKp44

Benyamin Rosental; Michael Brusilovsky; Uzi Hadad; Dafna Oz; Michael Y. Appel; Fabian Afergan; Rami Yossef; Lior Rosenberg; Amir Aharoni; Adelheid Cerwenka; Kerry S. Campbell; Alex Braiman; Angel Porgador

NK cells play an important role in the early immune response to cancer. The NKp44 activating receptor is the only natural cytotoxicity receptor that is expressed exclusively by primate NK cells, yet its cellular ligands remain largely unknown. Proliferating cell nuclear Ag (PCNA) is overexpressed in cancer cells. In this study, we show that the NKp44 receptor recognizes PCNA. Their interaction inhibits NK cell function through NKp44/ITIM. The physical interaction of NKp44 and PCNA is enabled by recruitment of target cell PCNA to the NK immunological synapse. We demonstrate that PCNA promotes cancer survival by immune evasion through inhibition of NKp44-mediated NK cell attack.


Journal of Proteome Research | 2009

Natural Cytotoxicity Receptors NKp30, NKp44 and NKp46 Bind to Different Heparan Sulfate/Heparin Sequences

Marie-Lyn Hecht; Benyamin Rosental; Tim Horlacher; Oren Hershkovitz; Jose L. de Paz; Christian Noti; Stefan Schauer; Angel Porgador; Peter H. Seeberger

Natural Killer (NK) cells recognize and destroy tumors and virus-infected cells in an antibody-independent manner. The regulation of NK cells is mediated by activating and inhibiting receptors on the NK cell surface. One important family of activating receptors is the natural cytotoxicity receptors (NCRs) which include NKp30, NKp44 and NKp46. The NCRs initiate tumor targeting by recognition of heparan sulfate on cancer cells. This study aims to elucidate heparan sulfate structural motifs that are important for NCR binding. Microarray and surface plasmon resonance experiments with a small library of heparan sulfate/heparin oligosaccharides helped to clarify the binding preferences of the three NCRs. We demonstrate that the NCRs interact with highly charged HS/heparin structures, but differ in preferred modification patterns and chain lengths. The affinity of NKp30 and NKp44 for synthetic HS/heparin is approximately one order of magnitude higher than the affinity of NKp46. We further show the relevance of synthetic HS/heparin for the binding of NCRs to tumor cells and for NCR-mediated activation of natural killer cells. In conclusion, NCRs recognize different microdomains on heparan sulfate with different affinities.


Journal of Immunology | 2013

Genome-wide siRNA screen reveals a new cellular partner of NK cell receptor KIR2DL4: heparan sulfate directly modulates KIR2DL4-mediated responses.

Michael Brusilovsky; Moti Cordoba; Benyamin Rosental; Oren Hershkovitz; Mark Andrake; Anna Pecherskaya; Margret B. Einarson; Yan Zhou; Alex Braiman; Kerry S. Campbell; Angel Porgador

KIR2DL4 (CD158d) is a distinct member of the killer cell Ig-like receptor (KIR) family in human NK cells that can induce cytokine production and cytolytic activity in resting NK cells. Soluble HLA-G, normally expressed only by fetal-derived trophoblast cells, was reported to be a ligand for KIR2DL4; however, KIR2DL4 expression is not restricted to the placenta and can be found in CD56high subset of peripheral blood NK cells. We demonstrated that KIR2DL4 can interact with alternative ligand(s), expressed by cells of epithelial or fibroblast origin. A genome-wide high-throughput siRNA screen revealed that KIR2DL4 recognition of cell-surface ligand(s) is directly regulated by heparan sulfate (HS) glucosamine 3-O-sulfotransferase 3B1 (HS3ST3B1). KIR2DL4 was found to directly interact with HS/heparin, and the D0 domain of KIR2DL4 was essential for this interaction. Accordingly, exogenous HS/heparin can regulate cytokine production by KIR2DL4-expressing NK cells and HEK293T cells (HEK293T-2DL4), and induces differential localization of KIR2DL4 to rab5+ and rab7+ endosomes, thus leading to downregulation of cytokine production and degradation of the receptor. Furthermore, we showed that intimate interaction of syndecan-4 (SDC4) HS proteoglycan (HSPG) and KIR2DL4 directly affects receptor endocytosis and membrane trafficking.


PLOS ONE | 2012

Glycans in Sera of Amyotrophic Lateral Sclerosis Patients and Their Role in Killing Neuronal Cells

Meital Edri-Brami; Benyamin Rosental; Dana Hayoun; Michael Welt; Hila Rosen; Itzhak Wirguin; Beatrice Nefussy; Vivian E. Drory; Angel Porgador; Rachel G. Lichtenstein

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease caused by degeneration of upper and lower motor neurons. To date, glycosylation patterns of glycoproteins in fluids of ALS patients have not been described. Moreover, the aberrant glycosylation related to the pathogenesis of other neurodegenerative diseases encouraged us to explore the glycome of ALS patient sera. We found high levels of sialylated glycans and low levels of core fucosylated glycans in serum-derived N-glycans of patients with ALS, compared to healthy volunteer sera. Based on these results, we analyzed the IgG Fc N297-glycans, as IgG are major serum glycoproteins affected by sialylation or core fucosylation and are found in the motor cortex of ALS patients. The analyses revealed a distinct glycan, A2BG2, in IgG derived from ALS patient sera (ALS-IgG). This glycan increases the affinity of IgG to CD16 on effector cells, consequently enhancing Antibody-Dependent Cellular Cytotoxicity (ADCC). Therefore, we explore whether the Fc-N297-glycans of IgG may be involved in ALS disease. Immunostaining of brain and spinal cord tissues revealed over-expression of CD16 and co-localization of intact ALS-IgG with CD16 and in brain with activated microglia of G93A-SOD1 mice. Intact ALS-IgG enhanced effector cell activation and ADCC reaction in comparison to sugar-depleted or control IgG. ALS-IgG were localized in the synapse between brain microglia and neurons of G93A-SOD1 mice, manifesting a promising in vivo ADCC reaction. Therefore, glycans of ALS-IgG may serve as a biomarker for the disease and may be involved in neuronal damage.


Current Medicinal Chemistry | 2012

The effect of chemotherapy/radiotherapy on cancerous pattern recognition by NK cells.

Benyamin Rosental; Michael Y. Appel; Rami Yossef; Uzi Hadad; Michael Brusilovsky; Angel Porgador

In recent years, the effects of cancer chemotherapy and radiotherapy (CT/RT) regimens as they apply to the immune system have been explored. NK cells represent the main cytotoxic arm of the innate immune system, and their functionality is vital to establishing an effective anti tumor immune response. This review examines current CT/RT interventions in light of their effects on NK cell functionality. The effects of CT/RT on the expression of the various ligands for activating and inhibitory NK cell receptors are discussed. Expression of ligands for the activating NKG2D receptor is enhanced by cell stress; accordingly there are numerous reports of their higher expression in cells exposed to various CT/RT agents. In contrast, some agents have been reported to cause ligand shedding, which can serve to inhibit NK cell activity. Reported effects of CT/RT on tumor expression of ligands for the activating Natural Cytotoxicity Receptors, and of HLA class I ligands for NK cell inhibitory receptors are also noted. Additionally, we describe reports concerning the direct effects of CT/RT on NK cell function. Many treatments adversely affect NK cell function directly, but observations made through in vitro systems may differ from those obtained utilizing clinical samples. The effects of CT/RT on both direct NK cell cytotoxicity and on NK cell-mediated Antibody Dependent Cellular Cytotoxicity are explored. Taken together, CT/RT affects NK cell anti-tumor immunity from multiple angles. The interplay is complex, and future work is needed to achieve the optimal synergy between CT/RT and innate as well as adaptive immunity in the treatment of cancer.


Journal of Immunotoxicology | 2012

Human NK cell recognition of target cells in the prism of natural cytotoxicity receptors and their ligands

Michael Brusilovsky; Benyamin Rosental; Avishai Shemesh; Michael Y. Appel; Angel Porgador

The matter of the pathogen- and cancer-associated ligands recognized by the Natural Cytotoxicity Receptors (NCRs) has been a subject of intense research ever since the identification of the NCRs more than 12 years ago by Alessandro and Lorenzo Moretta: NKp46 in 1997, NKp44 in 1998, and finally NKp30 in 1999. Expression patterns recognized by NCRs include pathogen-derived, pathogen-induced, and cancer-associated cellular ‘self’ ligands. Pathogen-exposed cells may exhibit both types of pathogen-associated ligands. Transformed cells, in contrast, exhibit only ‘self’ ligands which are derived from both the intracellular- and membrane-associated milieu of self molecules. These expression patterns allow for NCR-based NK cell discrimination between healthy and affected cells, in the realms of both pathogenic infection and potential tumorigenesis. The focus of this review is on the current knowledge regarding the identities of NCR ligands and the type of target cells expressing these ligands.


Journal of Immunology | 2012

Dimerization of NKp46 Receptor Is Essential for NKp46-Mediated Lysis: Characterization of the Dimerization Site by Epitope Mapping

Michal Jaron-Mendelson; Rami Yossef; Michael Y. Appel; Alon Zilka; Uzi Hadad; Fabian Afergan; Benyamin Rosental; Stanislav Engel; Shlomo Nedvetzki; Alex Braiman; Angel Porgador

NKp46 is a primary activating receptor of NK cells that is involved in lysis of target cells by NK cells. Previous studies showed that the membrane-proximal domain of NKp46 (NKp46D2) retained the binding of NKp46 to its ligands and is involved in lysis. We studied NKp46D2 by using a peptide-based epitope mapping approach and identified an NKp46D2-derived linear epitope that inhibited NKp46-mediated lysis. The epitope, designated as pep4 (aa 136–155), interacted with NKp46, and lysis by NK cells was inhibited by the presence of pep4. Through modeling and mutagenesis, we showed that pep4 could be involved in NKp46 homodimerization. R145 and D147 contribute to the function of pep4, and R145Q mutation in recombinant NKp46 reduced its binding to target cells. At the cellular level, fluorescent resonance energy transfer analysis revealed that pep4 is indeed involved in dimerization of cell membrane-associated NKp46. We suggest that the NKp46-derived pep4 site is part of the dimerization surface of NKp46 and that NKp46 dimerization contributes to NKp46-mediated lysis by NK cells.


Nature Immunology | 2018

Engagement of MHC class I by the inhibitory receptor LILRB1 suppresses macrophages and is a target of cancer immunotherapy

Kipp Weiskopf; Kevin S. Kao; Sydney R. Gordon; Benyamin Rosental; Ying Y. Yiu; Benson M. George; Maxim Markovic; Nan Guo Ring; Jonathan M. Tsai; Kelly M. McKenna; Po Yi Ho; Robin Z. Cheng; James Y. Chen; Layla J. Barkal; Aaron M. Ring; Irving L. Weissman; Roy L. Maute

Exciting progress in the field of cancer immunotherapy has renewed the urgency of the need for basic studies of immunoregulation in both adaptive cell lineages and innate cell lineages. Here we found a central role for major histocompatibility complex (MHC) class I in controlling the phagocytic function of macrophages. Our results demonstrated that expression of the common MHC class I component β2-microglobulin (β2M) by cancer cells directly protected them from phagocytosis. We further showed that this protection was mediated by the inhibitory receptor LILRB1, whose expression was upregulated on the surface of macrophages, including tumor-associated macrophages. Disruption of either MHC class I or LILRB1 potentiated phagocytosis of tumor cells both in vitro and in vivo, which defines the MHC class I–LILRB1 signaling axis as an important regulator of the effector function of innate immune cells, a potential biomarker for therapeutic response to agents directed against the signal-regulatory protein CD47 and a potential target of anti-cancer immunotherapy.Host cells display ‘don’t eat me’ signals to protect themselves from phagocytosis. Maute and colleagues identify a novel ‘don’t eat me’ system based on recognition of MHC class I by the phagocyte-expressed inhibitory molecule LILRB1.


Oncotarget | 2016

Survival in acute myeloid leukemia is associated with NKp44 splice variants.

Avishai Shemesh; Michael Brusilovsky; Uzi Hadad; Omri Teltsh; Avishay Edri; Eitan Rubin; Kerry S. Campbell; Benyamin Rosental; Angel Porgador

NKp44 is a receptor encoded by the NCR2 gene, which is expressed by cytokine-activated natural killer (NK) cells that are involved in anti-AML immunity. NKp44 has three splice variants corresponding to NKp44ITIM+ (NKp44-1) and NKp44ITIM− (NKp44-2, and NKp44-3) isoforms. RNAseq data of AML patients revealed similar survival of NKp46+NKp44+ and NKp46+NKp44− patients. However, if grouped according to the NKp44 splice variant profile, NKp44-1 expression was significantly associated with poor survival of AML patients. Moreover, activation of PBMC from healthy controls showed co-dominant expression of NKp44-1 and NKp44-3, while primary NK clones show more diverse NKp44 splice variant profiles. Cultured primary NK cells resulted in NKp44-1 dominance and impaired function associated with PCNA over-expression by target cells. This impaired functional phenotype could be rescued by blocking of NKp44 receptor. Human NK cell lines revealed co-dominant expression of NKp44-1 and NKp44-3 and showed a functional phenotype that was not inhibited by PCNA over-expression. Furthermore, transfection-based overexpression of NKp44-1, but not NKp44-2/NKp44-3, reversed the endogenous resistance of NK-92 cells to PCNA-mediated inhibition, and resulted in poor formation of stable lytic immune synapses. This research contributes to the understanding of AML prognosis by shedding new light on the functional implications of differential splicing of NKp44.

Collaboration


Dive into the Benyamin Rosental's collaboration.

Top Co-Authors

Avatar

Angel Porgador

Ben-Gurion University of the Negev

View shared research outputs
Top Co-Authors

Avatar

Michael Brusilovsky

Ben-Gurion University of the Negev

View shared research outputs
Top Co-Authors

Avatar

Uzi Hadad

Ben-Gurion University of the Negev

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael Y. Appel

Ben-Gurion University of the Negev

View shared research outputs
Top Co-Authors

Avatar

Oren Hershkovitz

Ben-Gurion University of the Negev

View shared research outputs
Top Co-Authors

Avatar

Alex Braiman

Ben-Gurion University of the Negev

View shared research outputs
Top Co-Authors

Avatar

Avishai Shemesh

Ben-Gurion University of the Negev

View shared research outputs
Top Co-Authors

Avatar

Rami Yossef

Ben-Gurion University of the Negev

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge