Bio


I graduated with a Ph.D from the laboratory of Prof. Eliahu Zlotkin at the Hebrew University of Jerusalem, where my research focused on Insect-selective neurotoxins expressing Baculoviruses. In December 2000 I joined the laboratory of Professor Jeffrey Glenn's at Stanford School of Medicine as a postdoctoral fellow. Here my research focused on HCV membrane association, specifically the association of NS5A and NS4B with host cell membranes and its role in HCV replication.
As a Senior Research Scientist at Professor Glenn's lab my interests focus around three major themes:
1. Broad spectrum antivirals targeting host functions
2. Viral genome RNA structures as antiviral targets
3. None Alcoholic Steatohepatitis (NASH) and its progression to hepatocellular carcinoma (HCC)

Current Role at Stanford


Senior Research Scientist

Education & Certifications


  • Ph.D., The Hebrew University of Jerusalem, Cell Biology (2000)
  • M.Sc., The Hebrew University of Jerusalem, Life Science (1994)
  • B.Sc., The Hebrew University of Jerusalem, Biology (1993)

All Publications


  • Programmable antivirals targeting critical conserved viral RNA secondary structures from influenza A virus and SARS-CoV-2. Nature medicine Hagey, R. J., Elazar, M., Pham, E. A., Tian, S., Ben-Avi, L., Bernardin-Souibgui, C., Yee, M. F., Moreira, F. R., Rabinovitch, M. V., Meganck, R. M., Fram, B., Beck, A., Gibson, S. A., Lam, G., Devera, J., Kladwang, W., Nguyen, K., Xiong, A., Schaffert, S., Avisar, T., Liu, P., Rustagi, A., Fichtenbaum, C. J., Pang, P. S., Khatri, P., Tseng, C., Taubenberger, J. K., Blish, C. A., Hurst, B. L., Sheahan, T. P., Das, R., Glenn, J. S. 2022

    Abstract

    Influenza A virus's (IAV's) frequent genetic changes challenge vaccine strategies and engender resistance to current drugs. We sought to identify conserved and essential RNA secondary structures within IAV's genome that are predicted to have greater constraints on mutation in response to therapeutic targeting. We identified and genetically validated an RNA structure (packaging stem-loop 2 (PSL2)) that mediates in vitro packaging and in vivo disease and is conserved across all known IAV isolates. A PSL2-targeting locked nucleic acid (LNA), administered 3d after, or 14d before, a lethal IAV inoculum provided 100% survival in mice, led to the development of strong immunity to rechallenge with a tenfold lethal inoculum, evaded attempts to select for resistance and retained full potency against neuraminidase inhibitor-resistant virus. Use of an analogous approach to target SARS-CoV-2, prophylactic administration of LNAs specific for highly conserved RNA structures in the viral genome, protected hamsters from efficient transmission of the SARS-CoV-2 USA_WA1/2020 variant. These findings highlight the potential applicability of this approach to any virus of interest via a process we term 'programmable antivirals', with implications for antiviral prophylaxis and post-exposure therapy.

    View details for DOI 10.1038/s41591-022-01908-x

    View details for PubMedID 35982307

  • Combination of Novel Therapies for HDV. Viruses Elazar, M., Glenn, J. S. 2022; 14 (2)

    Abstract

    Treatment options for HDV have been limited to interferon alfa-based therapies with its poor efficacy to side effects ratio. Several novel therapies have now advanced into the clinic. As they each have a different mechanism of action, there is the potential for combination therapy. Here we review how studying the HDV life cycle has led to the development of these novel therapies, the key developments leading to, and the details of, the first combination study of novel anti-HDV therapies, and suggest what additional combinations of novel therapies can be anticipated as we enter this exciting new area of HDV treatments.

    View details for DOI 10.3390/v14020268

    View details for PubMedID 35215860

  • Letter to the Editor regarding article "Emerging concepts for the treatment of hepatitis delta" [Menashe Elazar and Jeffrey S Glenn, Curr Opin Virol 24 (2017) 55-59] Reply CURRENT OPINION IN VIROLOGY Elazar, M., Glenn, J. 2018; 28: 169

    View details for PubMedID 29456115

  • Quantitative Evaluation of Viral Protein Binding to Phosphoinositide Receptors and Pharmacological Inhibition ANALYTICAL CHEMISTRY Kim, S., Jackman, J. A., Elazar, M., Cho, S., Glenn, J. S., Cho, N. 2017; 89 (18): 9742–50

    Abstract

    There is significant interest in developing analytical methods to characterize molecular recognition events between proteins and phosphoinositides, which are a medically important class of carbohydrate-functionalized lipids. Within this scope, one area of high priority involves quantitatively evaluating drug candidates that pharmacologically inhibit protein-phosphoinositide interactions. As full-length proteins are often difficult to produce, establishing methods to study these interactions with shorter, bioactive peptides would be advantageous. Herein, we report an atomic force microscopy (AFM)-based force spectroscopic approach to detect the specific interaction between an amphipathic, α-helical (AH) peptide derived from the hepatitis C virus NS5A protein and its biological target, the phosphatidylinositol (4,5)-bisphosphate [PI(4,5)P2] phosphoinositide receptor. After optimization of the peptide tethering strategy and measurement parameters, the binding specificity of AH peptide for PI(4,5)P2 receptors was comparatively evaluated across a panel of phosphoinositides and the influence of ionic strength on AH-PI(4,5)P2 binding strength was tested. Importantly, these capabilities were translated into the development of a novel experimental methodology to determine the inhibitory activity of a small-molecule drug candidate acting against the AH-PI(4,5)P2 interaction, and extracted kinetic parameters agree well with literature values obtained by conventional biochemical methods. Taken together, our findings provide a nanomechanical basis for explaining the high binding specificity of the NS5A AH to PI(4,5)P2 receptors, in turn establishing an analytical framework to study phosphoinositide-binding viral peptides and proteins as well as a broadly applicable approach to evaluate candidate inhibitors of protein-phosphoinositide interactions.

    View details for PubMedID 28809547

  • Long-term culture of human liver tissue with advanced hepatic functions. JCI insight Ng, S. S., Xiong, A., Nguyen, K., Masek, M., No, D. Y., Elazar, M., Shteyer, E., Winters, M. A., Voedisch, A., Shaw, K., Rashid, S. T., Frank, C. W., Cho, N. J., Glenn, J. S. 2017; 2 (11)

    Abstract

    A major challenge for studying authentic liver cell function and cell replacement therapies is that primary human hepatocytes rapidly lose their advanced function in conventional, 2-dimensional culture platforms. Here, we describe the fabrication of 3-dimensional hexagonally arrayed lobular human liver tissues inspired by the liver's natural architecture. The engineered liver tissues exhibit key features of advanced differentiation, such as human-specific cytochrome P450-mediated drug metabolism and the ability to support efficient infection with patient-derived inoculums of hepatitis C virus. The tissues permit the assessment of antiviral agents and maintain their advanced functions for over 5 months in culture. This extended functionality enabled the prediction of a fatal human-specific hepatotoxicity caused by fialuridine (FIAU), which had escaped detection by preclinical models and short-term clinical studies. The results obtained with the engineered human liver tissue in this study provide proof-of-concept determination of human-specific drug metabolism, demonstrate the ability to support infection with human hepatitis virus derived from an infected patient and subsequent antiviral drug testing against said infection, and facilitate detection of human-specific drug hepatotoxicity associated with late-onset liver failure. Looking forward, the scalability and biocompatibility of the scaffold are also ideal for future cell replacement therapeutic strategies.

    View details for DOI 10.1172/jci.insight.90853

    View details for PubMedID 28570275

  • Hepatitis delta infection - Current and new treatment options BEST PRACTICE & RESEARCH CLINICAL GASTROENTEROLOGY Elazar, M., Koh, C., Glenn, J. S. 2017; 31 (3): 321–27

    Abstract

    In humans, hepatitis D virus (HDV) infection only occurs in the presence of a concomitant hepatitis B virus (HBV) infection, and induces the most severe form of human viral hepatitis. Even though HDV is spread worldwide and is endemic in some regions, screening and treatment has been often neglected in part due to the lack of an effective therapy. Moreover, HDV prevalence rates are increasing in many countries driven by immigration from areas of high endemicity. Currently, no FDA-approved anti-HDV therapy is available, although interferon (IFN) alpha therapy has demonstrated benefit in a minority of patients. In this review, we present a current view of our understanding of the epidemiology, molecular virology and management of HDV infection. We additionally discuss new treatment approaches in development and describe the most promising results of recent and ongoing clinical trials of these new potential agents.

    View details for PubMedID 28774414

  • Emerging concepts for the treatment of hepatitis delta. Current opinion in virology Elazar, M., Glenn, J. S. 2017; 24: 55-59

    Abstract

    Hepatitis delta virus (HDV) causes the most severe form of human viral hepatitis and is associated with a higher risk of cirrhosis, liver decompensation and liver cancer. Interferon alpha is the only agent that has demonstrated efficacy to date, although response rates are low and it is associated with significant side effects. A better understanding of the relevant molecular virology has resulted in the identification of new candidate targets. Future therapeutic options are rapidly evolving as several new agents have entered clinical development, including the entry inhibitor myrcludex-B, the nucleic acid polymer REP2139-Ca inhibiting HBV surface antigen secretion, the farnesyltransferase inhibitor lonafarnib that targets virus assembly, and a better tolerated interferon-interferon lambda.

    View details for DOI 10.1016/j.coviro.2017.04.004

    View details for PubMedID 28475945

  • A novel quantitative microarray antibody capture (Q-MAC) assay identifies an extremely high HDV prevalence amongst HBV infected Mongolians. Hepatology Chen, X., Oidovsambuu, O., Liu, P., Grosely, R., Elazar, M., Winn, V. D., Fram, B., Boa, Z., Dai, H., Dashtseren, B., Yagaanbuyant, D., Genden, Z., Dashdorj, N., Bungert, A., Dashdorj, N., Glenn, J. S. 2016

    Abstract

    Hepatitis delta virus (HDV) causes the most severe form of human viral hepatitis. HDV requires a hepatitis B virus (HBV) co-infection to provide HDV with HBV surface antigen envelope proteins. The net effect of HDV is to make the underlying HBV disease worse, including higher rates of hepatocellular carcinoma (HCC). Accurate assessments of current HDV prevalence have been hampered by the lack of readily available and reliable quantitative assays, combined with the absence of an FDA-approved therapy. We sought to develop a convenient assay for accurately screening populations and to use this assay to determine HDV prevalence in a population with abnormally high rates of HCC. We developed a high throughput quantitative microarray antibody capture (Q-MAC) assay for anti-HDV IgG wherein recombinant HDV delta antigen is printed by microarray on slides coated with a noncontinuous, nanostructured plasmonic gold film, enabling quantitative fluorescent detection of anti-HDV antibody in small aliquots of patient serum. This assay was then used to screen all HBV-infected patients identified in a large randomly selected cohort designed to represent the Mongolian population. We identified two quantitative thresholds of captured antibody that were 100% predictive of the sample either being positive on standard western blot, or harboring HDV RNA detectable by qPCR, respectively. Subsequent screening of the HBV-positive cohort revealed that a remarkable 57% were RNA positive and an additional 4% were positive on western blot alone.The Q-MAC assay's unique performance characteristics make it ideal for population screening. Its application to the Mongolian HBsAg+ population reveals an apparent ∼60% prevalence of HDV co-infection amongst these HBV-infected Mongolian subjects, which may help explain the extraordinarily high rate of HCC in Mongolia. This article is protected by copyright. All rights reserved.

    View details for DOI 10.1002/hep.28957

    View details for PubMedID 27880976

  • Phosphatidylinositol 4,5-Bisphosphate Is an HCV NS5A Ligand and Mediates Replication of the Viral Genome. Gastroenterology Cho, N., Lee, C., Pang, P. S., Pham, E. A., Fram, B., Nguyen, K., Xiong, A., Sklan, E. H., Elazar, M., Koytak, E. S., Kersten, C., Kanazawa, K. K., Frank, C. W., Glenn, J. S. 2015; 148 (3): 616-625

    Abstract

    Phosphoinositides (PIs) bind and regulate localization of proteins via a variety of structural motifs. PI 4,5-bisphosphate (PI[4,5]P2) interacts with and modulates the function of several proteins involved in intracellular vesicular membrane trafficking. We investigated interactions between PI(4,5)P2 and hepatitis C virus (HCV) nonstructural protein 5A (NS5A) and effects on the viral life cycle.We used a combination of quartz crystal microbalance, circular dichroism, molecular genetics, and immunofluorescence to study specific binding of PI(4,5)P2 by the HCV NS5A protein. We evaluated the effects of PI(4,5)P2 on the function of NS5A by expressing wild-type or mutant forms of Bart79I or FL-J6/JFH-5'C19Rluc2AUbi21 RNA in Huh7 cells. We also studied the effects of strategies designed to inhibit PI(4,5)P2 on HCV replication in these cells.The N-terminal amphipathic helix of NS5A bound specifically to PI(4,5)P2, inducing a conformational change that stabilized the interaction between NS5A and TBC1D20, which is required for HCV replication. A pair of positively charged residues within the amphipathic helix (the basic amino acid PI(4,5)P2 pincer domain) was required for PI(4,5)P2 binding and replication of the HCV-RNA genome. A similar motif was found to be conserved across all HCV isolates, as well as amphipathic helices of many pathogens and apolipoproteins.PI(4,5)P2 binds to HCV NS5A to promote replication of the viral RNA genome in hepatocytes. Strategies to disrupt this interaction might be developed to inhibit replication of HCV and other viruses.

    View details for DOI 10.1053/j.gastro.2014.11.043

    View details for PubMedID 25479136

  • The interaction between the Hepatitis C proteins NS4B and NS5A is involved in viral replication. Virology David, N., Yaffe, Y., Hagoel, L., Elazar, M., Glenn, J. S., Hirschberg, K., Sklan, E. H. 2015; 475: 139-149

    Abstract

    Hepatitis C virus (HCV) replicates in membrane associated, highly ordered replication complexes (RCs). These complexes include viral and host proteins necessary for viral RNA genome replication. The interaction network among viral and host proteins underlying the formation of these RCs is yet to be thoroughly characterized. Here, we investigated the association between NS4B and NS5A, two critical RC components. We characterized the interaction between these proteins using fluorescence resonance energy transfer and a mammalian two-hybrid system. Specific tryptophan residues within the C-terminal domain (CTD) of NS4B were shown to mediate this interaction. Domain I of NS5A, was sufficient to mediate its interaction with NS4B. Mutations in the NS4B CTD tryptophan residues abolished viral replication. Moreover, one of these mutations also affected NS5A hyperphosphorylation. These findings provide new insights into the importance of the NS4B-NS5A interaction and serve as a starting point for studying the complex interactions between the replicase subunits.

    View details for DOI 10.1016/j.virol.2014.10.021

    View details for PubMedID 25462354

  • HCV NS5A Inhibitors: The Devil Is in the Details GASTROENTEROLOGY Elazar, M., Glenn, J. S. 2014; 147 (2): 273–77

    View details for PubMedID 24976028

  • The Anti-Genomic (Negative) Strand of Hepatitis C Virus Is Not Targetable by shRNA Lisowski, L., Elazar, M., Chu, K., Glenn, J. S., Kay, M. A. NATURE PUBLISHING GROUP. 2013: S75
  • The anti-genomic (negative) strand of Hepatitis C Virus is not targetable by shRNA. Nucleic acids research Lisowski, L., Elazar, M., Chu, K., Glenn, J. S., Kay, M. A. 2013; 41 (6): 3688-3698

    Abstract

    Hepatitis C Virus (HCV) and other plus-strand RNA viruses typically require the generation of a small number of negative genomes (20-100× lower than the positive genomes) for replication, making the less-abundant antigenome an attractive target for RNA interference(RNAi)-based therapy. Because of the complementarity of duplex short hairpin RNA/small interfering RNA (shRNA/siRNAs) with both genomic and anti-genomic viral RNA strands, and the potential of both shRNA strands to become part of the targeting complexes, preclinical RNAi studies cannot distinguish which viral strand is actually targeted in infected cells. Here, we addressed the question whether the negative HCV genome was bioaccessible to RNAi. We first screened for the most active shRNA molecules against the most conserved regions in the HCV genome, which were then used to generate asymmetric anti-HCV shRNAs that produce biologically active RNAi specifically directed against the genomic or antigenomic HCV sequences. Using this simple but powerful and effective method to screen for shRNA strand selectivity, we demonstrate that the antigenomic strand of HCV is not a viable RNAi target during HCV replication. These findings provide new insights into HCV biology and have important implications for the design of more effective and safer antiviral RNAi strategies seeking to target HCV and other viruses with similar replicative strategies.

    View details for DOI 10.1093/nar/gkt068

    View details for PubMedID 23396439

  • Using Chimeric Mice with Humanized Livers to Predict Human Drug Metabolism and a Drug-Drug Interaction JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS Nishimura, T., Hu, Y., Wu, M., Pham, E., Suemizu, H., Elazar, M., Liu, M., Idilman, R., Yurdaydin, C., Angus, P., Stedman, C., Murphy, B., Glenn, J., Nakamura, M., Nomura, T., Chen, Y., Zheng, M., Fitch, W. L., Peltz, G. 2013; 344 (2): 388-396

    Abstract

    Interspecies differences in drug metabolism have made it difficult to use preclinical animal testing data to predict the drug metabolites or potential drug-drug interactions (DDIs) that will occur in humans. Although chimeric mice with humanized livers can produce known human metabolites for test substrates, we do not know whether chimeric mice can be used to prospectively predict human drug metabolism or a possible DDI. Therefore, we investigated whether they could provide a more predictive assessment for clemizole, a drug in clinical development for the treatment of hepatitis C virus (HCV) infection. Our results demonstrate, for the first time, that analyses performed in chimeric mice can correctly identify the predominant human drug metabolite before human testing. The differences in the rodent and human pathways for clemizole metabolism were of importance, because the predominant human metabolite was found to have synergistic anti-HCV activity. Moreover, studies in chimeric mice also correctly predicted that a DDI would occur in humans when clemizole was coadministered with a CYP3A4 inhibitor. These results demonstrate that using chimeric mice can improve the quality of preclinical drug assessment.

    View details for DOI 10.1124/jpet.112.198697

    View details for PubMedID 23143674

  • Structural Map of a MicroRNA-122: Hepatitis C Virus Complex JOURNAL OF VIROLOGY Pang, P. S., Pham, E. A., Elazar, M., Patel, S. G., Eckart, M. R., Glenn, J. S. 2012; 86 (2): 1250-1254

    Abstract

    MicroRNA-122 (miR-122) enhances hepatitis C virus (HCV) fitness via targeting two sites in the 5'-untranslated region (UTR) of HCV. We used selective 2'-hydroxyl acylation analyzed by primer extension to resolve the HCV 5'-UTR's RNA secondary structure in the presence of miR-122. Nearly all nucleotides in miR-122 are involved in targeting the second site, beyond classic seed base pairings. These additional interactions enhance HCV replication in cell culture. To our knowledge, this is the first biophysical study of this complex to reveal the importance of 'tail' miR-122 nucleotide interactions.

    View details for DOI 10.1128/JVI.06367-11

    View details for PubMedID 22072754

  • Simplified RNA secondary structure mapping by automation of SHAPE data analysis NUCLEIC ACIDS RESEARCH Pang, P. S., Elazar, M., Pham, E. A., Glenn, J. S. 2011; 39 (22)

    Abstract

    SHAPE (Selective 2'-hydroxyl acylation analysed by primer extension) technology has emerged as one of the leading methods of determining RNA secondary structure at the nucleotide level. A significant bottleneck in using SHAPE is the complex and time-consuming data processing that is required. We present here a modified data collection method and a series of algorithms, embodied in a program entitled Fast Analysis of SHAPE traces (FAST), which significantly reduces processing time. We have used this method to resolve the secondary structure of the first ~900 nt of the hepatitis C virus (HCV) genome, including the entire core gene. We have also demonstrated the ability of SHAPE/FAST to detect the binding of a small molecule inhibitor to the HCV internal ribosomal entry site (IRES). In conclusion, FAST allows for high-throughput data processing to match the current high-throughput generation of data possible with SHAPE, reducing the barrier to determining the structure of RNAs of interest.

    View details for DOI 10.1093/nar/gkr773

    View details for PubMedID 21965531

  • Using 'Humanized' TK-NOG mice to predict human drug metabolism and drug-drug interactions 17th North American Regional International-Society-for-the-Study-of-Xenobiotics (ISSX) Meeting Hu, Y., Nishimura, T., Wu, M., Suemizu, H., Elazar, M., Glenn, J., Hasegawa, M., Nakamura, M., Nomura, T., Chen, Y., Zheng, M., Fitch, W. L., Peltz, G. INFORMA HEALTHCARE. 2011: 164–165
  • NOVEL ANTI-HCV THERAPY: SINGLE SHRNA TARGETING BOTH STRANDS OF HCV Lisowski, L., Elazar, M., Chu, K., Glenn, J. S., Kay, M. A. WILEY-BLACKWELL. 2011: 418–19
  • The hepatitis C virus NS5A inhibitor (BMS-790052) alters the subcellular localization of the NS5A non-structural viral protein VIROLOGY Lee, C., Ma, H., Hang, J. Q., Leveque, V., Sklan, E. H., Elazar, M., Klumpp, K., Glenn, J. S. 2011; 414 (1): 10-18

    Abstract

    The hepatitis C virus (HCV) non-structural (NS) 5A protein plays an essential role in the replication of the viral RNA by the membrane-associated replication complex (RC). Recently, a putative NS5A inhibitor, BMS-790052, exhibited the highest potency of any known anti-HCV compound in inhibiting HCV replication in vitro and showed a promising clinical effect in HCV-infected patients. The precise mechanism of action for this new class of potential anti-HCV therapeutics, however, is still unclear. In order to gain further insight into its mode of action, we sought to test the hypothesis that the antiviral effect of BMS-790052 might be mediated by interfering with the functional assembly of the HCV RC. We observed that BMS-790052 indeed altered the subcellular localization and biochemical fractionation of NS5A. Taken together, our data suggest that NS5A inhibitors such as BMS-790052 can suppress viral genome replication by altering the proper localization of NS5A into functional RCs.

    View details for DOI 10.1016/j.virol.2011.03.026

    View details for PubMedID 21513964

  • NS4B Targets and Inhibitors HEPATITIS C: ANTIVIRAL DRUG DISCOVERY AND DEVELOPMENT Elazar, M., Glenn, J. S., Tan, S. L., He, Y. P. 2011: 257–69
  • Identification of a Class of HCV Inhibitors Directed Against the Nonstructural Protein NS4B SCIENCE TRANSLATIONAL MEDICINE Cho, N., Dvory-Sobol, H., Lee, C., Cho, S., Bryson, P., Masek, M., Elazar, M., Frank, C. W., Glenn, J. S. 2010; 2 (15)

    Abstract

    New classes of drugs are needed to combat hepatitis C virus (HCV), an important worldwide cause of liver disease. We describe an activity of a key domain, an amphipathic helix we termed 4BAH2, within a specific HCV nonstructural protein, NS4B. In addition to its proposed role in viral replication, we validate 4BAH2 as essential for HCV genome replication and identify first-generation small-molecule inhibitors of 4BAH2 that specifically prevent HCV replication within cells. Mechanistic studies reveal that the inhibitors target 4BAH2 function by preventing either 4BAH2 oligomerization or 4BAH2 membrane association. 4BAH2 inhibitors represent an additional class of compounds with potential to effectively treat HCV.

    View details for DOI 10.1126/scitranslmed.3000331

    View details for PubMedID 20371471

  • The Anti-Hepatitis C Agent Nitazoxanide Induces Phosphorylation of Eukaryotic Initiation Factor 2 alpha Via Protein Kinase Activated by Double-Stranded RNA Activation GASTROENTEROLOGY Elazar, M., Liu, M., Mckenna, S. A., Liu, P., Gehrig, E. A., Puglisi, J. D., Rossignol, J., Glenn, J. S. 2009; 137 (5): 1827-1835

    Abstract

    New therapies are needed to treat patients infected with hepatitis C virus (HCV), a major worldwide cause of chronic liver disease. Nitazoxanide (NTZ), originally used to treat cryptosporidiosis infection, recently was shown to have unexpected antiviral activity in the HCV replicon system and in chronically infected patients. A pilot clinical study suggested that NTZ can augment the antiviral effect of interferon (IFN), although the molecular basis for its effect was unknown.We analyzed the effects of NTZ on the regulation of eukaryotic initiation factor-2alpha (eIF2alpha) and its IFN-induced kinase, protein kinase activated by double-stranded RNA (PKR), in cells that support HCV RNA replication and in vitro biochemical assays.NTZ increased eIF2alpha phosphorylation, a modification known to mediate host cell antiviral defenses. The addition of IFN to cell cultures increased NTZ-induced eIF2alpha phosphorylation. NTZ also increased PKR phosphorylation. In vitro, NTZ promoted PKR autophosphorylation, a key step in activating PKR's kinase activity for eIF2alpha. Finally, NTZ-induced eIF2alpha phosphorylation was reduced in the presence of specific inhibitors of PKR autophosphorylation.An important mechanism of NTZ's action involves activation of PKR, a key kinase that regulates the cell's innate antiviral response. These observations could explain the clinical antiviral effect of NTZ. NTZ might represent a new class of small molecules capable of potentiating and recapitulating important antiviral effects of IFN.

    View details for DOI 10.1053/j.gastro.2009.07.056

    View details for PubMedID 19664635

  • AAV Based RNAi Therapies To Treat and/or Prevent HCV in Animal Models Lisowski, L., Elazar, M., Grompe, M., Glenn, J. S., Kay, M. A. NATURE PUBLISHING GROUP. 2009: S14
  • A Role for Nitazoxanide in Combination with STAT-C Agents for Inhibition of HCV Replication and the Potential for the Prevention of Viral Resistance Korba, B., Glenn, J., Elazar, M., Rossignol, J. ELSEVIER SCIENCE BV. 2009: A20
  • Viral infection of human progenitor and liver-derived cells encapsulated in three-dimensional PEG-based hydrogel BIOMEDICAL MATERIALS Cho, N., Elazar, M., Xiong, A., Lee, W., Chiao, E., Baker, J., Frank, C. W., Glenn, J. S. 2009; 4 (1)

    Abstract

    We have studied the encapsulation of human progenitor cells into 3D PEG hydrogels. Replication-incompetent lentivirus promoter reporter vectors were found to efficiently detect the in vivo expression of human hepatic genes in hydrogel-encapsulated liver progenitor cells. Similarly, hydrogel-encapsulated cells could be efficiently infected with hepatitis C virus, and progeny infectious virus could be recovered from the media supernatants of the hydrogels. Provocatively, the diameters of these virus particles range from approximately 50 to 100 nm, while the calculated mesh size of the 8 k hydrogel is 44.6 +/- 1.7 A. To reconcile how viral particles can penetrate the hydrogels to infect the encapsulated cells, we propose that microfractures/defects of the hydrogel result in a functional pore size of up to 20 fold greater than predicted by theoretical mesh calculations. These results suggest a new model of hydrogel structure, and have exciting implications for tissue engineering and hepatitis virus studies.

    View details for DOI 10.1088/1748-6041/4/1/011001

    View details for PubMedID 18981544

  • Potential for Hepatitis C Virus Resistance to Nitazoxanide or Tizoxanide ANTIMICROBIAL AGENTS AND CHEMOTHERAPY Korba, B. E., Elazar, M., Lui, P., Rossignol, J., Glenn, J. S. 2008; 52 (11): 4069-4071

    Abstract

    Nitazoxanide and its primary metabolite, tizoxanide, inhibit hepatitis C virus (HCV) replication in HCV replicon systems. To study the potential for resistance, we subjected Huh7 cells harboring HCV replicons to serial passage in 250 muM G418 and increasing concentrations of nitazoxanide or tizoxanide. Passage of the replicon-containing cell lines in either compound resulted in increases in the 50% effective concentrations (EC(50)s) (7- to 13-fold), EC(90)s (14- to 36-fold), and 50% cytotoxic concentrations (2- to 4-fold) of both compounds. Serial passage in either compound did not alter the susceptibility of HCV replicons to ribavirin or 2'-C-methylcytidine. Interestingly, serial passage in nitazoxanide or tizoxanide resulted in increased sensitivity to alpha interferon 2b: EC(50)s and EC(90)s were reduced three- and eightfold, respectively. Replicons isolated from these cell lines had no greater ability to confer tizoxanide resistance, or increased susceptibility to alpha interferon, than replicons isolated from the parental cell line that had not previously been exposed to nitazoxanide or tizoxanide. These findings are indicative of a cell-mediated activity differing from that of other anti-HCV drugs but complementary with interferon and are consistent with the enhanced response rates observed clinically when nitazoxanide is combined with pegylated interferon therapy. Finally, unlike data for other compounds in advanced clinical development for HCV, these data are consistent with resistance in HCV replicon-containing cell lines conferred by changes in the host and not by mutations in the virus.

    View details for DOI 10.1128/AAC.00078-08

    View details for Web of Science ID 000260305600035

    View details for PubMedID 18710916

    View details for PubMedCentralID PMC2573111

  • PHARMACOLOGICAL INHIBITORS OF A NEW HEPATITIS C TARGET-RNA BINDING BY NS4B-DISCOVERED BY MICROFLUIDIC AFFINITY ANALYSIS 59th Annual Meeting of the American-Association-for-the-Study-of-Liver-Diseases Einav, S., Gerber, D., Bryson, P. D., Sklan, E., Elazar, M., Maerkl, S., Dvory, H. S., Machlin, E., Gu, W., Quake, S., Glenn, J. S. WILEY-BLACKWELL. 2008: 356A–356A
  • POTENTIAL ROLE FOR NITAZOXANIDE IN COMBINATION WITH STAT-C AGENTS FOR THE INHIBITION OF HCV REPLICATION WITHOUT THE DEVELOPMENT OF RESISTANCE Korba, B., Elazar, M., Liu, P., Glenn, J. S., Rossignol, J. JOHN WILEY & SONS INC. 2008: 356A
  • Discovery of a hepatitis C target and its pharmacological inhibitors by microfluidic affinity analysis NATURE BIOTECHNOLOGY Einav, S., Gerber, D., Bryson, P. D., Sklan, E. H., Elazar, M., Maerkl, S. J., Glenn, J. S., Quake, S. R. 2008; 26 (9): 1019-1027

    Abstract

    More effective therapies are urgently needed against hepatitis C virus (HCV), a major cause of viral hepatitis. We used in vitro protein expression and microfluidic affinity analysis to study RNA binding by the HCV transmembrane protein NS4B, which plays an essential role in HCV RNA replication. We show that HCV NS4B binds RNA and that this binding is specific for the 3' terminus of the negative strand of the viral genome with a dissociation constant (Kd) of approximately 3.4 nM. A high-throughput microfluidic screen of a compound library identified 18 compounds that substantially inhibited binding of RNA by NS4B. One of these compounds, clemizole hydrochloride, was found to inhibit HCV RNA replication in cell culture that was mediated by its suppression of NS4B's RNA binding, with little toxicity for the host cell. These results yield new insight into the HCV life cycle and provide a candidate compound for pharmaceutical development.

    View details for DOI 10.1038/nbt.1490

    View details for PubMedID 18758449

  • Isolation and transcriptional profiling of purified hepatic cells derived from human embryonic stem cells STEM CELLS Chiao, E., Elazar, M., Xing, Y., Xiong, A., Kmet, M., Millan, M. T., Glenn, J. S., Wong, W. H., Baker, J. 2008; 26 (8): 2032-2041

    Abstract

    The differentiation of human embryonic stem cells (hESCs) into functional hepatocytes provides a powerful in vitro model system for studying the molecular mechanisms governing liver development. Furthermore, a well-characterized renewable supply of hepatocytes differentiated from hESCs could be used for in vitro assays of drug metabolism and toxicology, screening of potential antiviral agents, and cell-based therapies to treat liver disease. In this study, we describe a protocol for the differentiation of hESCs toward hepatic cells with complex cellular morphologies. Putative hepatic cells were identified and isolated using a lentiviral vector, containing the alpha-fetoprotein promoter driving enhanced green fluorescent protein expression (AFP:eGFP). Whole-genome transcriptional profiling was performed on triplicate samples of AFP:eGFP+ and AFP:eGFP- cell populations using the recently released Affymetrix Exon Array ST 1.0 (Santa Clara, CA, http://www.affymetrix.com). Statistical analysis of the transcriptional profiles demonstrated that the AFP:eGFP+ population is highly enriched for genes characteristic of hepatic cells. These data provide a unique insight into the complex process of hepatocyte differentiation, point to signaling pathways that may be manipulated to more efficiently direct the differentiation of hESCs toward mature hepatocytes, and identify molecular markers that may be used for further dissection of hepatic cell differentiation from hESCs. Disclosure of potential conflicts of interest is found at the end of this article.

    View details for DOI 10.1634/stemcells.2007-0964

    View details for PubMedID 18535157

  • A Rab-GAP TBC domain protein binds hepatitis C virus NS5A and mediates viral replication JOURNAL OF VIROLOGY Sklan, E. H., Staschke, K., Oakes, T. M., Elazar, M., Winters, M., Aroeti, B., Danieli, T., Glenn, J. S. 2007; 81 (20): 11096-11105

    Abstract

    Hepatitis C virus (HCV) is an important cause of liver disease worldwide. Current therapies are inadequate for most patients. Using a two-hybrid screen, we isolated a novel cellular binding partner interacting with the N terminus of HCV nonstructural protein NS5A. This partner contains a TBC Rab-GAP (GTPase-activating protein) homology domain found in all known Rab-activating proteins. As the first described interaction between such a Rab-GAP and a viral protein, this finding suggests a new mechanism whereby viruses may subvert host cell machinery for mediating the endocytosis, trafficking, and sorting of their own proteins. Moreover, depleting the expression of this partner severely impairs HCV RNA replication with no obvious effect on cell viability. These results suggest that pharmacologic disruption of this NS5A-interacting partner can be contemplated as a potential new antiviral strategy against a pathogen affecting nearly 3% of the world's population.

    View details for DOI 10.1128/JVI.01249-07

    View details for PubMedID 17686842

  • Bj alpha IT: a novel scorpion alpha-toxin selective for insects - unique pharmacological tool INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY Arnon, T., Potikha, T., Sher, D., Elazar, M., Mao, W. F., Tal, T., Bosmans, F., Tytgat, J., Ben-Arie, N., Zlotkin, E. 2005; 35 (3): 187–95

    Abstract

    Long-chain neurotoxins derived from the venom of the Buthidae scorpions, which affect voltage-gated sodium channels (VGSCs) can be subdivided according to their toxicity to insects into insect-selective excitatory and depressant toxins (beta-toxins) and the alpha-like toxins which affect both mammals and insects. In the present study by the aid of reverse-phase HPLC column chromatography, RT-PCR, cloning and various toxicity assays, a new insect selective toxin designated as BjalphaIT was isolated from the venom of the Judean Black Scorpion (Buthotus judaicus), and its full primary sequence was determined: MNYLVVICFALLLMTVVESGRDAYIADNLNCAYTCGSNSYCNTECTKNGAVSGYCQWLGKYGNACWCINLPDKVPIRIPGACR (leader sequence is underlined). Despite its lack of toxicity to mammals and potent toxicity to insects, BjalphaIT reveals an amino acid sequence and an inferred spatial arrangement that is characteristic of the well-known scorpion alpha-toxins highly toxic to mammals. BjalphaITs sharp distinction between insects and mammals was also revealed by its effect on sodium conductance of two cloned neuronal VGSCs heterloguously expressed in Xenopus laevis oocytes and assayed with the two-electrode voltage-clamp technique. BjalphaIT completely inhibits the inactivation process of the insect para/tipE VGSC at a concentration of 100 nM, in contrast to the rat brain Na(v)1.2/beta1 which is resistant to the toxin. The above categorical distinction between mammal and insect VGSCs exhibited by BjalphaIT enables its employment in the clarification of the molecular basis of the animal group specificity of scorpion venom derived neurotoxic polypeptides and voltage-gated sodium channels.

    View details for PubMedID 15705498

  • A nucleotide binding motif in hepatitis C virus (HCV) NS4B mediates HCV RNA replication JOURNAL OF VIROLOGY Einav, S., Elazar, M., Danieli, T., Glenn, J. S. 2004; 78 (20): 11288-11295

    Abstract

    Hepatitis C virus (HCV) is a major cause of viral hepatitis. There is no effective therapy for most patients. We have identified a nucleotide binding motif (NBM) in one of the virus's nonstructural proteins, NS4B. This structural motif binds and hydrolyzes GTP and is conserved across HCV isolates. Genetically disrupting the NBM impairs GTP binding and hydrolysis and dramatically inhibits HCV RNA replication. These results have exciting implications for the HCV life cycle and novel antiviral strategies.

    View details for PubMedID 15452248

  • An n-terminal amphipathic helix in hepatitis C virus (HCV) NS4B mediates membrane association, correct localization of replication complex proteins, and HCV RNA replication JOURNAL OF VIROLOGY Elazar, M., Liu, P., Rice, C. M., Glenn, J. S. 2004; 78 (20): 11393-11400

    Abstract

    Like other positive-strand RNA viruses, hepatitis C virus (HCV) is believed to replicate its RNA in association with host cell cytoplasmic membranes. Because of its association with such membranes, NS4B, one of the virus's nonstructural proteins, may play an important role in this process, although the mechanistic details are not well understood. We identified a putative N-terminal amphipathic helix (AH) in NS4B that mediates membrane association. Introduction of site-directed mutations designed to disrupt the hydrophobic face of the AH abolishes the AH's ability to mediate membrane association. An AH in NS4B is conserved across HCV isolates. Completely disrupting the amphipathic nature of NS4B's N-terminal helix abolished HCV RNA replication, whereas partial disruption resulted in an intermediate level of replication. Finally, immunofluorescence studies revealed that HCV replication complex components were mislocalized in the AH-disrupted mutant. These results identify a key membrane-targeting domain which can form the basis for developing novel antiviral strategies.

    View details for DOI 10.1128/JVI.78.20.11393-11400.2004

    View details for PubMedID 15452261

  • Amphipathic helix-dependent localization of NS5A mediates hepatitis C virus RNA replication JOURNAL OF VIROLOGY Elazar, M., Cheong, K. H., Liu, P., Greenberg, H. B., Rice, C. M., Glenn, J. S. 2003; 77 (10): 6055-6061

    Abstract

    We identified an N-terminal amphipathic helix (AH) in one of hepatitis C virus (HCV)'s nonstructural proteins, NS5A. This AH is necessary and sufficient for membrane localization and is conserved across isolates. Genetically disrupting the AH impairs HCV replication. Moreover, an AH peptide-mimic inhibits the membrane association of NS5A in a dose-dependent manner. These results have exciting implications for the HCV life cycle and novel antiviral strategies.

    View details for DOI 10.1128/JVI.77.10.6055-6061.2003

    View details for PubMedID 12719597

  • The pharmacologic versatility of a neurotoxic polypeptide. Postepy higieny i medycyny doswiadczalnej Elazar, M., Shichor, I., Zlotkin, E. 2002; 56 (3): 411-20

    View details for PubMedID 12194254

  • Targeting of an expressed neurotoxin by its recombinant baculovirus JOURNAL OF EXPERIMENTAL BIOLOGY Elazar, M., Levi, R., Zlotkin, E. 2001; 204 (15): 2637–45

    Abstract

    AaIT, an insect-selective neurotoxic polypeptide derived from scorpion venom, has recently been used to engineer recombinant baculoviruses for insect pest control. Lepidopterous larvae infected with an AaIT-expressing baculovirus reveal symptoms of paralysis identical to those induced by injection of the native toxin. However, the paralyzed larvae treated by the recombinant virus possess an approximately 50-fold lower hemolymph toxin concentration than insects paralyzed by the native toxin. The mechanism of this potentiation effect was studied using immunocytochemistry, electrophysiology and toxicity assays. (i) Light microscopy, using peroxidase-conjugated antibodies, revealed the presence of toxin in virus-susceptible tissues, including tracheal epithelia located close to the central nervous system and beyond its lamellar enveloping sheath. (ii) High-resolution immunogold electron microscopical cytochemistry clearly revealed the presence of recombinant AaIT toxin inside the thoracic and abdominal ganglia on neuronal cell bodies and axonal membranes. (iii) Ventral nerve cords dissected from silkworm larvae infected with the recombinant baculovirus exhibited a high degree of excitability, expressed as enhanced frequency and bursting mode of their spontaneous activity, when compared to nerve cords infected with the wild-type virus. We conclude that the recombinant-virus-infected tracheal epithelia, outbranching in the body of an infected insect, (i) locally supply a continuous, freshly produced toxin to its neuronal receptors and (ii) introduce the expressed toxin to the insect central nervous system, thus providing it with critical target sites that are inaccessible to the native toxin.

    View details for Web of Science ID 000170563500004

    View details for PubMedID 11533113

  • AaIT: From neurotoxin to insecticide BIOCHIMIE Zlotkin, E., Fishman, Y., Elazar, M. 2000; 82 (9-10): 869–81

    Abstract

    AaIT is a single chain neurotoxic polypeptide derived from the venom of the Buthid scorpion Androctonus australis Hector, composed of 70 amino acids cross-linked by four disulfide bridges. Its strict selectivity for insects has been documented by toxicity, electrophysiological and ligand receptor binding assays. These last have shown that various insect neuronal membranes possess a single class of non-interacting AaIT binding sites of high affinity (K(D) = 1-3(n)M) and low capacity (0.5-2.0 pmol/mg prot.). The fast excitatory paralysis induced by AaIT is a result of a presynaptic effect, namely the induction of a repetitive firing in the terminal branches of the insect's motor nerves resulting in a massive and uncoordinated stimulation of the respective skeletal muscles. The neuronal repetitive activity is attributed to an exclusive and specific perturbation of sodium conductance as a consequence of toxin binding to external loops of the insect voltage-dependent sodium channel and modification of its gating mechanism. From a strictly agrotechnical point of view AaIT involvement in plant protection has taken the following two complementary forms: firstly, as a factor for the genetic engineering of insect infective baculoviruses resulting in potent and selective bio-insecticides. The efficacy of the AaIT-expressing, recombinant baculovirus is attributed mainly to its ability to continuously provide and translocate the gene of the expressed toxin to the insect central nervous system; secondly, based on the pharmacological flexibility of the voltage-gated sodium channel, as a device for insecticide resistance management. Channel mutations conferring resistance to a given class of insecticidal agents (such as the KDR phenomenon) may greatly increase susceptibility to the AaIT expressing bioinsecticides. Thus the AaIT is a pharmacological tool for the study of insect neuronal excitability and chemical ecology and the development of new approaches to insect control.

    View details for DOI 10.1016/S0300-9084(00)01177-9

    View details for Web of Science ID 000165789900009

    View details for PubMedID 11086217