Network


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

Hotspot


Dive into the research topics where Elad Horwitz is active.

Publication


Featured researches published by Elad Horwitz.


Nature Immunology | 2008

Human microRNAs regulate stress-induced immune responses mediated by the receptor NKG2D

Noam Stern-Ginossar; Chamutal Gur; Moshe Biton; Elad Horwitz; Moran Elboim; Noa Stanietsky; Michal Mandelboim; Ofer Mandelboim

MICA and MICB are stress-induced ligands recognized by the activating receptor NKG2D. A microRNA encoded by human cytomegalovirus downregulates MICB expression by targeting a specific site in the MICB 3′ untranslated region. As this site is conserved among different MICB alleles and a similar site exists in the MICA 3′ untranslated region, we speculated that these sites are targeted by cellular microRNAs. Here we identified microRNAs that bound to these MICA and MICB 3′ untranslated region sequences and obtained data suggesting that these microRNAs maintain expression of MICA and MICB protein under a certain threshold and facilitate acute upregulation of MICA and MICB during cellular stress. These microRNAs were overexpressed in various tumors and we demonstrate here that they aided tumor avoidance of immune recognition.


Nature Immunology | 2011

Epithelial microRNAs regulate gut mucosal immunity via epithelium-T cell crosstalk

Moshe Biton; Avi Levin; Michal Slyper; Irit Alkalay; Elad Horwitz; Hagar Mor; Sharon Kredo-Russo; Tali Avnit-Sagi; Gady Cojocaru; Farid Zreik; Zvi Bentwich; Matthew N. Poy; David Artis; Michael D. Walker; Eran Hornstein; Eli Pikarsky; Yinon Ben-Neriah

Colonic homeostasis entails epithelium-lymphocyte cooperation, yet many participants in this process are unknown. We show here that epithelial microRNAs mediate the mucosa–immune system crosstalk necessary for mounting protective T helper type 2 (TH2) responses. Abolishing the induction of microRNA by gut-specific deletion of Dicer1 (Dicer1Δgut), which encodes an enzyme involved in microRNA biogenesis, deprived goblet cells of RELMβ, a key TH2 antiparasitic cytokine; this predisposed the host to parasite infection. Infection of Dicer1Δgut mice with helminths favored a futile TH1 response with hallmarks of inflammatory bowel disease. Interleukin 13 (IL-13) induced the microRNA miR-375, which regulates the expression of TSLP, a TH2-facilitating epithelial cytokine; this indicated a TH2-amplification loop. We found that miR-375 was required for RELMβ expression in vivo; miR-375-deficient mice had significantly less intestinal RELMβ, which possibly explains the greater susceptibility of Dicer1Δgut mice to parasites. Our findings indicate that epithelial microRNAs are key regulators of gut homeostasis and mucosal immunity.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Mutant KRAS is a druggable target for pancreatic cancer

Elina Zorde Khvalevsky; Racheli Gabai; Itzhak Haim Rachmut; Elad Horwitz; Zivia Brunschwig; Ariel Orbach; Adva Shemi; Talia Golan; Abraham J. Domb; Eylon Yavin; Hilla Giladi; Ludmila Rivkin; Alina Simerzin; Rami Eliakim; Abed Khalaileh; Ayala Hubert; Maor Lahav; Yael Kopelman; Eran Goldin; Alan Dancour; Yael Hants; Sagit Arbel-Alon; Rinat Abramovitch; Amotz Shemi; Eithan Galun

Significance Pancreatic cancer is still one of the major challenges in clinical oncology. Mutant KRAS is a driving oncogene in the majority of human pancreatic cancer cases. We have made an effort to meet this challenge by developing a therapeutic platform for local and prolonged delivery of siRNA. Our results show that the siRNA targeted against KRAS mutations with a local prolonged release system knocks down KRAS expression in vitro and in vivo, leading to an antitumor effect. Our report describes an applicable and efficient delivery method of siRNA that overcomes the major obstacles of toxicity and organ accessibility. Notably our approach enabled the conversion of KRAS from a nondruggable to a potentially druggable cancer target. Pancreatic ductal adenocarcinoma (PDA) represents an unmet therapeutic challenge. PDA is addicted to the activity of the mutated KRAS oncogene which is considered so far an undruggable therapeutic target. We propose an approach to target KRAS effectively in patients using RNA interference. To meet this challenge, we have developed a local prolonged siRNA delivery system (Local Drug EluteR, LODER) shedding siRNA against the mutated KRAS (siG12D LODER). The siG12D LODER was assessed for its structural, release, and delivery properties in vitro and in vivo. The effect of the siG12D LODER on tumor growth was assessed in s.c. and orthotopic mouse models. KRAS silencing effect was further assessed on the KRAS downstream signaling pathway. The LODER-encapsulated siRNA was stable and active in vivo for 155 d. Treatment of PDA cells with siG12D LODER resulted in a significant decrease in KRAS levels, leading to inhibition of proliferation and epithelial–mesenchymal transition. In vivo, siG12D LODER impeded the growth of human pancreatic tumor cells and prolonged mouse survival. We report a reproducible and safe delivery platform based on a miniature biodegradable polymeric matrix, for the controlled and prolonged delivery of siRNA. This technology provides the following advantages: (i) siRNA is protected from degradation; (ii) the siRNA is slowly released locally within the tumor for prolonged periods; and (iii) the siG12D LODER elicits a therapeutic effect, thereby demonstrating that mutated KRAS is indeed a druggable target.


Hepatology | 2009

S100A8 and S100A9 are novel nuclear factor kappa B target genes during malignant progression of murine and human liver carcinogenesis

Julia Németh; Ilan Stein; Daniel Haag; Astrid Riehl; Thomas Longerich; Elad Horwitz; Kai Breuhahn; Christoffer Gebhardt; Peter Schirmacher; Meinhard Hahn; Yinon Ben-Neriah; Eli Pikarsky; Peter Angel; Jochen Hess

The nuclear factor‐kappaB (NF‐κB) signaling pathway has been recently shown to participate in inflammation‐induced cancer progression. Here, we describe a detailed analysis of the NF‐κB–dependent gene regulatory network in the well‐established Mdr2 knockout mouse model of inflammation‐associated liver carcinogenesis. Expression profiling of NF‐κB–deficient and NF‐κB–proficient hepatocellular carcinoma (HCC) revealed a comprehensive list of known and novel putative NF‐κB target genes, including S100a8 and S100a9. We detected increased co‐expression of S100A8 and S100A9 proteins in mouse HCC cells, in human HCC tissue, and in the HCC cell line Hep3B on ectopic RelA expression. Finally, we found a synergistic function for S100A8 and S100A9 in Hep3B cells resulting in a significant induction of reactive oxygen species (ROS), accompanied by enhanced cell survival. Conclusion: We identified S100A8 and S100A9 as novel NF‐κB target genes in HCC cells during inflammation‐associated liver carcinogenesis and provide experimental evidence that increased co‐expression of both proteins supports malignant progression by activation of ROS‐dependent signaling pathways and protection from cell death. (HEPATOLOGY 2009.)


Gut | 2012

NKp46-mediated killing of human and mouse hepatic stellate cells attenuates liver fibrosis

Chamutal Gur; S. Doron; Shlomit Kfir-Erenfeld; Elad Horwitz; Lina Abu-Tair; Rifaat Safadi; Ofer Mandelboim

Background Liver fibrosis, which involves activation of hepatic stellate cells (HSC), is a major health problem and is the end outcome of all chronic liver diseases. The liver is populated with lymphocytes, among which are natural killer (NK) cells, whose activity is controlled by inhibitory and activating receptors. NKp46, one of the major NK activating receptors expressed by NK cells, is also a specific NK marker that discriminates NK cells from all other lymphocyte subsets. It recognises viral haemagglutinins and unknown cellular ligands. Methods The anti-fibrotic activity of the NKp46 receptor was assessed in vivo and in vitro using NKp46-deficient mice (NCR1gfp/gfp), the carbon tetrachloride model and in vitro NK killing assays. Primary murine and human HSC were stained for the expression of the NKp46 ligand using fusion proteins composed of the extracellular portions of the murine and human NKp46 receptors fused to human IgG1. Results It was shown that murine HSC express a ligand for the murine orthologue of the NKp46 receptor, NCR1. NCR1 inhibited liver fibrosis in vivo; in vitro, murine HSC were killed in an NCR1-dependent manner. In humans it was shown that human HSC also express a ligand for the human NKp46 receptor and that the killing of human HSC is NKp46 dependent. Conclusions In addition to NKG2D, NKp46/NCR1 play an important role in inhibition of liver fibrosis. This suggests that fibrosis can be better controlled through the manipulation of NKp46 activity.


Cancer Discovery | 2014

Human and Mouse VEGFA-Amplified Hepatocellular Carcinomas Are Highly Sensitive to Sorafenib Treatment

Elad Horwitz; Ilan Stein; Mariacarla Andreozzi; Julia Németh; Avivit Shoham; Orit Pappo; Nora Schweitzer; Luigi Tornillo; Naama Kanarek; Luca Quagliata; Farid Zreik; Rinnat M. Porat; Rutie Finkelstein; Hendrik Reuter; Ronald Koschny; Tom M. Ganten; Carolin Mogler; Oren Shibolet; Jochen Hess; Kai Breuhahn; Myriam Grunewald; Peter Schirmacher; Arndt Vogel; Luigi Terracciano; Peter Angel; Yinon Ben-Neriah; Eli Pikarsky

UNLABELLED Death rates from hepatocellular carcinoma (HCC) are steadily increasing, yet therapeutic options for advanced HCC are limited. We identify a subset of mouse and human HCCs harboring VEGFA genomic amplification, displaying distinct biologic characteristics. Unlike common tumor amplifications, this one seems to work via heterotypic paracrine interactions; stromal VEGF receptors (VEGFR), responding to tumor VEGF-A, produce hepatocyte growth factor (HGF) that reciprocally affects tumor cells. VEGF-A inhibition results in HGF downregulation and reduced proliferation, specifically in amplicon-positive mouse HCCs. Sorafenib-the first-line drug in advanced HCC-targets multiple kinases, including VEGFRs, but has only an overall mild beneficial effect. We found that VEGFA amplification specifies mouse and human HCCs that are distinctly sensitive to sorafenib. FISH analysis of a retrospective patient cohort showed markedly improved survival of sorafenib-treated patients with VEGFA-amplified HCCs, suggesting that VEGFA amplification is a potential biomarker for HCC response to VEGF-A-blocking drugs. SIGNIFICANCE Using a mouse model of inflammation-driven cancer, we identified a subclass of HCC carrying VEGFA amplification, which is particularly sensitive to VEGF-A inhibition. We found that a similar amplification in human HCC identifies patients who favorably responded to sorafenib-the first-line treatment of advanced HCC-which has an overall moderate therapeutic efficacy.


Nature Medicine | 2016

p16Ink4a-induced senescence of pancreatic beta cells enhances insulin secretion

Aharon Helman; Agnes Klochendler; Narmen Azazmeh; Yael Gabai; Elad Horwitz; Shira Anzi; Avital Swisa; Reba Condiotti; Roy Z. Granit; Yuval Nevo; Yaakov Fixler; Dorin Shreibman; Amit Zamir; Sharona Tornovsky-Babeay; Chunhua Dai; Benjamin Glaser; Alvin C. Powers; A. M. James Shapiro; Mark A. Magnuson; Yuval Dor; Ittai Ben-Porath

Cellular senescence is thought to contribute to age-associated deterioration of tissue physiology. The senescence effector p16Ink4a is expressed in pancreatic beta cells during aging and limits their proliferative potential; however, its effects on beta cell function are poorly characterized. We found that beta cell–specific activation of p16Ink4a in transgenic mice enhances glucose-stimulated insulin secretion (GSIS). In mice with diabetes, this leads to improved glucose homeostasis, providing an unexpected functional benefit. Expression of p16Ink4a in beta cells induces hallmarks of senescence—including cell enlargement, and greater glucose uptake and mitochondrial activity—which promote increased insulin secretion. GSIS increases during the normal aging of mice and is driven by elevated p16Ink4a activity. We found that islets from human adults contain p16Ink4a-expressing senescent beta cells and that senescence induced by p16Ink4a in a human beta cell line increases insulin secretion in a manner dependent, in part, on the activity of the mechanistic target of rapamycin (mTOR) and the peroxisome proliferator-activated receptor (PPAR)-γ proteins. Our findings reveal a novel role for p16Ink4a and cellular senescence in promoting insulin secretion by beta cells and in regulating normal functional tissue maturation with age.


Genes & Development | 2010

Spermatogenesis rescue in a mouse deficient for the ubiquitin ligase SCFβ-TrCP by single substrate depletion

Naama Kanarek; Elad Horwitz; Inbal Mayan; Michael Leshets; Gady Cojocaru; Matti Davis; Ben-Zion Tsuberi; Eli Pikarsky; Michele Pagano; Yinon Ben-Neriah

beta-TrCP, the substrate recognition subunit of a Skp1-Cul1-F-box (SCF) ubiquitin ligase, is ubiquitously expressed from two distinct paralogs, targeting many regulatory proteins for proteasomal degradation. We generated inducible beta-TrCP hypomorphic mice and found that they are surprisingly healthy, yet have a severe testicular defect. We show that the two beta-TrCP paralogs have a nonredundant role in spermatogenesis. The testicular defect is tightly associated with cell adhesion failure within the seminiferous tubules and is fully reversible upon beta-TrCP restoration. Remarkably, testicular depletion of a single beta-TrCP substrate, Snail1, rescued the adhesion defect and restored spermatogenesis. Our studies highlight an unexpected functional reserve of this central E3, as well as a bottleneck in a specific tissue: a single substrate whose stabilization is incompatible with testicular differentiation.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Critical role for IL-1β in DNA damage-induced mucositis

Naama Kanarek; Sergei I. Grivennikov; Michael Leshets; Audrey Lasry; Irit Alkalay; Elad Horwitz; Yoav D. Shaul; Matthew D. Stachler; Elena Voronov; Ron N. Apte; Michele Pagano; Eli Pikarsky; Michael Karin; Sankar Ghosh; Yinon Ben-Neriah

Significance Deletion of the E3 β-TrCP in the mouse gut epithelium deregulates enterocyte cell cycle, induces a DNA damage response (DDR), and abolishes the epithelium barrier function, resulting in a lethal mucosal inflammation. Epithelial-derived IL-1β, likely induced by DDR independently of NF-κB, is a major culprit, and initiates the pathology by compromising epithelial tight junctions (TJs). Anti–IL-1β treatment secures the TJs and prevents the fulminant mucosal inflammation. IL-1β secretion accompanies human mucositis, a severe mucosal inflammatory reaction caused by chemoradiation therapy-induced DNA damage, which often results in treatment suspension. We propose that anti–IL-1β preventive treatment may ameliorate mucositis, as well as multiple disorders associated with epithelial barrier permeability, including burn injuries, head and neck trauma, alcoholic intoxication, and graft-vs.-host disease. β-TrCP, the substrate recognition subunit of SCF-type ubiquitin ligases, is ubiquitously expressed from two distinct paralogs, targeting for degradation many regulatory proteins, among which is the NF-κB inhibitor IκB. To appreciate tissue-specific roles of β-TrCP, we studied the consequences of inducible ablation of three or all four alleles of the E3 in the mouse gut. The ablation resulted in mucositis, a destructive gut mucosal inflammation, which is a common complication of different cancer therapies and represents a major obstacle to successful chemoradiation therapy. We identified epithelial-derived IL-1β as the culprit of mucositis onset, inducing mucosal barrier breach. Surprisingly, epithelial IL-1β is induced by DNA damage via an NF-κB–independent mechanism. Tissue damage caused by gut barrier disruption is exacerbated in the absence of NF-κB, with failure to express the endogenous IL-1β receptor antagonist IL-1Ra upon four-allele loss. Antibody neutralization of IL-1β prevents epithelial tight junction dysfunction and alleviates mucositis in β-TrCP–deficient mice. IL-1β antagonists should thus be considered for prevention and treatment of severe morbidity associated with mucositis.


Hepatology | 2016

PI3K/AKT/mTOR-dependent stabilization of oncogenic far-upstream element binding proteins in hepatocellular carcinoma cells.

Jana Samarin; Vibor Laketa; Mona Malz; Stephanie Roessler; Ilan Stein; Elad Horwitz; Stephan Singer; Eleni Dimou; Antonio Cigliano; Michaela Bissinger; Christine S. Falk; Xin Chen; Steven Dooley; Eli Pikarsky; Diego F. Calvisi; Carsten Schultz; Peter Schirmacher; Kai Breuhahn

Transcription factors of the far‐upstream element‐binding protein (FBP) family represent cellular pathway hubs, and their overexpression in liver cancer (hepatocellular carcinoma [HCC]) stimulates tumor cell proliferation and correlates with poor prognosis. Here we determine the mode of oncogenic FBP overexpression in HCC cells. Using perturbation approaches (kinase inhibitors, small interfering RNAs) and a novel system for rapalog‐dependent activation of AKT isoforms, we demonstrate that activity of the phosphatidylinositol‐4,5‐biphosphate 3‐kinase/AKT pathway is involved in the enrichment of nuclear FBP1 and FBP2 in liver cancer cells. In human HCC tissues, phospho‐AKT significantly correlates with nuclear FBP1/2 accumulation and expression of the proliferation marker KI67. Mechanistic target of rapamycin (mTOR) inhibition or blockade of its downstream effector eukaryotic translation initiation factor 4E activity equally reduced FBP1/2 concentrations. The mTORC1 inhibitor rapamycin diminishes FBP enrichment in liver tumors after hydrodynamic gene delivery of AKT plasmids. In addition, the multikinase inhibitor sorafenib significantly reduces FBP levels in HCC cells and in multidrug resistance 2‐deficient mice that develop HCC due to severe inflammation. Both FBP1/2 messenger RNAs are highly stable, with FBP2 being more stable than FBP1. Importantly, inhibition of phosphatidylinositol‐4,5‐biphosphate 3‐kinase/AKT/mTOR signaling significantly diminishes FBP1/2 protein stability in a caspase‐3/‐7‐dependent manner. Conclusion: These data provide insight into a transcription‐independent mechanism of FBP protein enrichment in liver cancer; further studies will have to show whether this previously unknown interaction between phosphatidylinositol‐4,5‐biphosphate 3‐kinase/AKT/mTOR pathway activity and caspase‐mediated FBP stabilization allows the establishment of interventional strategies in FBP‐positive HCCs. (Hepatology 2016;63:813–826)

Collaboration


Dive into the Elad Horwitz's collaboration.

Top Co-Authors

Avatar

Eli Pikarsky

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Yinon Ben-Neriah

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Ilan Stein

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Yuval Dor

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Benjamin Glaser

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Ofer Mandelboim

Hebrew University of Jerusalem

View shared research outputs
Top Co-Authors

Avatar

Jochen Hess

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Julia Németh

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Kai Breuhahn

University Hospital Heidelberg

View shared research outputs
Top Co-Authors

Avatar

Peter Angel

German Cancer Research Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge