Network


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

Hotspot


Dive into the research topics where Julie Dyall is active.

Publication


Featured researches published by Julie Dyall.


Antimicrobial Agents and Chemotherapy | 2014

Repurposing of Clinically Developed Drugs for Treatment of Middle East Respiratory Syndrome Coronavirus Infection

Julie Dyall; Christopher M. Coleman; Brit J. Hart; Thiagarajan Venkataraman; Jason Kindrachuk; Reed F. Johnson; Gene G. Olinger; Peter B. Jahrling; Monique Laidlaw; Lisa M. Johansen; Calli M. Lear-Rooney; Pamela J. Glass; Lisa E. Hensley; Matthew B. Frieman

ABSTRACT Outbreaks of emerging infections present health professionals with the unique challenge of trying to select appropriate pharmacologic treatments in the clinic with little time available for drug testing and development. Typically, clinicians are left with general supportive care and often untested convalescent-phase plasma as available treatment options. Repurposing of approved pharmaceutical drugs for new indications presents an attractive alternative to clinicians, researchers, public health agencies, drug developers, and funding agencies. Given the development times and manufacturing requirements for new products, repurposing of existing drugs is likely the only solution for outbreaks due to emerging viruses. In the studies described here, a library of 290 compounds was screened for antiviral activity against Middle East respiratory syndrome coronavirus (MERS-CoV) and severe acute respiratory syndrome coronavirus (SARS-CoV). Selection of compounds for inclusion in the library was dependent on current or previous FDA approval or advanced clinical development. Some drugs that had a well-defined cellular pathway as target were included. In total, 27 compounds with activity against both MERS-CoV and SARS-CoV were identified. The compounds belong to 13 different classes of pharmaceuticals, including inhibitors of estrogen receptors used for cancer treatment and inhibitors of dopamine receptor used as antipsychotics. The drugs identified in these screens provide new targets for in vivo studies as well as incorporation into ongoing clinical studies.


Journal of General Virology | 2014

Interferon-β and mycophenolic acid are potent inhibitors of Middle East respiratory syndrome coronavirus in cell-based assays.

Brit J. Hart; Julie Dyall; Elena Postnikova; Huanying Zhou; Jason Kindrachuk; Reed F. Johnson; Gene G. Olinger; Matthew B. Frieman; Peter B. Jahrling; Lisa E. Hensley

The Middle East respiratory syndrome coronavirus (MERS-CoV) presents a novel emerging threat to public health worldwide. Several treatments for infected individuals have been suggested including IFN, ribavirin and passive immunotherapy with convalescent plasma. Administration of IFN-α2b and ribavirin has improved outcomes of MERS-CoV infection in rhesus macaques when administered within 8 h post-challenge. However, detailed and systematic evidence on the activity of other clinically available drugs is limited. Here we compared the susceptibility of MERS-CoV with different IFN products (IFN-α2b, IFN-γ, IFN-universal, IFN-α2a and IFN-β), as well as with two antivirals, ribavirin and mycophenolic acid (MPA), against MERS-CoV (Hu/Jordan-N3/2012) in vitro. Of all the IFNs tested, IFN-β showed the strongst inhibition of MERS-CoV in vitro, with an IC₅₀ of 1.37 U ml(-1), 41 times lower than the previously reported IC₅₀ (56.08 U ml(-1)) of IFN-α2b. IFN-β inhibition was confirmed in the virus yield reduction assay, with an IC90 of 38.8 U ml(-1). Ribavirin did not inhibit viral replication in vitro at a dose that would be applicable to current treatment protocols in humans. In contrast, MPA showed strong inhibition, with an IC₅₀ of 2.87 µM. This drug has not been previously tested against MERS-CoV and may provide an alternative to ribavirin for treatment of MERS-CoV. In conclusion, IFN-β, MPA or a combination of the two may be beneficial in the treatment of MERS-CoV or as a post-exposure intervention in high-risk patients with known exposures to MERS-CoV.


Journal of Virology | 2011

Comparative Analysis of Monkeypox Virus Infection of Cynomolgus Macaques by the Intravenous or Intrabronchial Inoculation Route

Reed F. Johnson; Julie Dyall; Dan R. Ragland; Louis Huzella; Russell Byrum; Catherine Jett; Marisa St. Claire; Alvin L. Smith; Jason Paragas; Joseph E. Blaney; Peter B. Jahrling

ABSTRACT Monkeypox virus (MPXV) infection has recently expanded in geographic distribution and can be fatal in up to 10% of cases. The intravenous (i.v.) inoculation of nonhuman primates (NHPs) results in an accelerated fulminant disease course compared to that of naturally occurring MPXV infection in humans. Alternative routes of inoculation are being investigated to define an NHP model of infection that more closely resembles natural disease progression. Our goal was to determine if the intrabronchial (i.b.) exposure of NHPs to MPXV results in a systemic disease that better resembles the progression of human MPXV infection. Here, we compared the disease course following an i.v. or i.b. inoculation of NHPs with 10-fold serial doses of MPXV Zaire. Classical pox-like disease was observed in NHPs administered a high virus dose by either route. Several key events were delayed in the highest doses tested of the i.b. model compared to the timing of the i.v. model, including the onset of fever, lesion appearance, peak viremia, viral shedding in nasal and oral swabs, peak cytokine levels, and time to reach endpoint criteria. Virus distribution across 19 tissues was largely unaffected by the inoculation route at the highest doses tested. The NHPs inoculated by the i.b. route developed a viral pneumonia that likely exacerbated disease progression. Based on the observations of the delayed onset of clinical and virological parameters and endpoint criteria that may more closely resemble those of human MPXV infection, the i.b. MPXV model should be considered for the further investigation of viral pathogenesis and countermeasures.


Antiviral Research | 2016

The lipid moiety of brincidofovir is required for in vitro antiviral activity against Ebola virus.

Laura K. McMullan; Mike Flint; Julie Dyall; César G. Albariño; Gene G. Olinger; Scott Foster; Phiroze Sethna; Lisa E. Hensley; Stuart T. Nichol; E. Randall Lanier; Christina F. Spiropoulou

Brincidofovir (BCV) is the 3-hexadecyloxy-1-propanol (HDP) lipid conjugate of the acyclic nucleoside phosphonate cidofovir (CDV). BCV has established broad-spectrum activity against double-stranded DNA (dsDNA) viruses; however, its activity against RNA viruses has been less thoroughly evaluated. Here, we report that BCV inhibited infection of Ebola virus in multiple human cell lines. Unlike the mechanism of action for BCV against cytomegalovirus and other dsDNA viruses, phosphorylation of CDV to the diphosphate form appeared unnecessary. Instead, antiviral activity required the lipid moiety and in vitro activity against EBOV was observed for several HDP-nucleotide conjugates.


Antimicrobial Agents and Chemotherapy | 2015

Antiviral Potential of ERK/MAPK and PI3K/AKT/mTOR Signaling Modulation for Middle East Respiratory Syndrome Coronavirus Infection as Identified by Temporal Kinome Analysis

Jason Kindrachuk; Britini L. Ork; Brit J. Hart; Steven Mazur; Matthew B. Frieman; Dawn Traynor; Reed F. Johnson; Julie Dyall; Jens H. Kuhn; Gene G. Olinger; Lisa E. Hensley; Peter B. Jahrling

ABSTRACT Middle East respiratory syndrome coronavirus (MERS-CoV) is a lineage C betacoronavirus, and infections with this virus can result in acute respiratory syndrome with renal failure. Globally, MERS-CoV has been responsible for 877 laboratory-confirmed infections, including 317 deaths, since September 2012. As there is a paucity of information regarding the molecular pathogenesis associated with this virus or the identities of novel antiviral drug targets, we performed temporal kinome analysis on human hepatocytes infected with the Erasmus isolate of MERS-CoV with peptide kinome arrays. bioinformatics analysis of our kinome data, including pathway overrepresentation analysis (ORA) and functional network analysis, suggested that extracellular signal-regulated kinase (ERK)/mitogen-activated protein kinase (MAPK) and phosphoinositol 3-kinase (PI3K)/serine-threonine kinase (AKT)/mammalian target of rapamycin (mTOR) signaling responses were specifically modulated in response to MERS-CoV infection in vitro throughout the course of infection. The overrepresentation of specific intermediates within these pathways determined by pathway and functional network analysis of our kinome data correlated with similar patterns of phosphorylation determined through Western blot array analysis. In addition, analysis of the effects of specific kinase inhibitors on MERS-CoV infection in tissue culture models confirmed these cellular response observations. Further, we have demonstrated that a subset of licensed kinase inhibitors targeting the ERK/MAPK and PI3K/AKT/mTOR pathways significantly inhibited MERS-CoV replication in vitro whether they were added before or after viral infection. Taken together, our data suggest that ERK/MAPK and PI3K/AKT/mTOR signaling responses play important roles in MERS-CoV infection and may represent novel drug targets for therapeutic intervention strategies.


The Journal of Infectious Diseases | 2011

Evaluation of Monkeypox Disease Progression by Molecular Imaging

Julie Dyall; Reed F. Johnson; Dar-Yeong Chen; Louis Huzella; Dan R. Ragland; Daniel J. Mollura; Russell Byrum; Richard C. Reba; Gerald Jennings; Peter B. Jahrling; Joseph E. Blaney; Jason Paragas

Infection of nonhuman primates (NHPs) with monkeypox virus (MPXV) is currently being developed as an animal model of variola infection in humans. We used positron emission tomography and computed tomography (PET/CT) to identify inflammatory patterns as predictors for the outcome of MPXV disease in NHPs. Two NHPs were sublethally inoculated by the intravenous (IV) or intrabronchial (IB) routes and imaged sequentially using fluorine-18 fluorodeoxyglucose ((18)FDG) uptake as a nonspecific marker of inflammation/immune activation. Inflammation was observed in the lungs of IB-infected NHPs, and bilobular involvement was associated with morbidity. Lymphadenopathy and immune activation in the axillary lymph nodes were evident in IV- and IB-infected NHPs. Interestingly, the surviving NHPs had significant (18)FDG uptake in the axillary lymph nodes at the time of MPXV challenge with no clinical signs of illness, suggesting an association between preexisting immune activation and survival. Molecular imaging identified patterns of inflammation/immune activation that may allow risk assessment of monkeypox disease.


Emerging Infectious Diseases | 2015

Lack of Effect of Lamivudine on Ebola Virus Replication

Lisa E. Hensley; Julie Dyall; Gene G. Olinger; Peter B. Jahrling

To the Editor: The unprecedented number of Ebola virus disease (EVD) cases in western Africa has compelled the world to consider experimental and off-label therapeutics to mitigate the current outbreak. For clinicians, approved drugs are an attractive solution because of known safety profiles and availability. Oral lamivudine (GlaxoSmithKline, Brentford, UK), a US Food and Drug Administration–approved anti-HIV drug, has been suggested as a possible antiviral agent against Ebola virus (EBOV). In September 2014, a Liberian physician, Dr. Gorbee Logan, reported positive results while treating EVD with lamivudine (1). Thirteen of 15 patients treated with lamivudine survived presumed EVD and were declared virus free. Clinical confirmation of EVD in these cases remains to be verified. Our laboratory had previously assessed this antiretroviral compound in drug screens against EBOV and observed no discernable antiviral activity. However, given the recent testimonials regarding lamivudine effectiveness in treating EBOV-infected patients in Africa, we conducted additional studies to determine whether our previous assertion that lamivudine lacked any direct antiviral activity was correct. Lamivudine is a nucleoside analog reverse transcription inhibitor of HIV and hepatitis B virus that acts as a synthetic cytidine analog. Incorporation of the active triphosphate form into viral DNA results in chain termination. Studies have demonstrated that lamivudine is a weak inhibitor of mammalian α, β, and γ DNA polymerases (2). Lamivudine would not be expected to inhibit the replication of a negative-strand RNA virus. The activity of lamivudine against EBOV infection was evaluated in a cell-based ELISA with 1995 isolate EBOV H. sapiens-tc/COD/1995/Kikwit (EBOV/Kik) (3). Three cell lines were tested: Vero E6 (African green monkey kidney, ATCC CRL-1586), Hep G2 (human hepatoma, ATCC HB-8065), and human monocyte-derived macrophages. Macrophages were generated by treating CD14+ cells for 7 days with macrophage colony–stimulating factor and conditioned medium. Cells were treated with compounds in 3-, 4-, or 8- point dose response curves with 2-fold dilutions starting at 80 µmol/L or 320 µmol/L oral lamivudine. Toremifene (T7204–5MG; Sigma-Aldrich, St. Louis, MO, USA) was used as a positive control for activity against EBOV and tested at 2-fold dilutions starting at 25 µmol/L. One hour after drug addition, the cells were infected at a multiplicity of infection of 0.5 or 1 with EBOV/Kik. Experiments were run on duplicate plates or the entire experiment was run on 2 separate days. At 48 hours after infection, cells were formalin-fixed and stained with a primary antibody against EBOV (antibodies against viral matrix protein or glycoprotein) and a secondary antibody (Alexa-488 or horseradish peroxidase). No direct antiviral effect for lamivudine was observed at concentrations ≤320 µmol/L in Vero E6 cells (Table). Because optimal efficacy of the drug requires phosphorylation, lack of activity may be caused by poor phosphorylation in Vero E6 cells (6). Therefore, we also assessed HepG2 cells and primary human monocyte–derived macrophages sensitive to EBOV infection. Toremifene was included as a positive control. Toremifene is a US Food and Drug Administration–approved drug that was reported to have direct antiviral activity in cell culture and to protect mice infected with mouse-adapted EBOV (3). As expected, toremifene inhibited EBOV at low micromolar concentrations (Table). Table Inhibitory effects of test compounds on Ebola virus replication* Finally, we assessed the antiviral activity of the compounds against a recent isolate prototype from the current outbreak, EBOV H. sapiens-tc/GIN/2014/Gueckedou-C05 (EBOV/Gue) to test whether inhibition of EBOV/Gue by lamivudine was different from that of the reference Kikwit strain. In contrast to a known active compound (toremifene), lamivudine showed no direct antiviral activity. The current data suggest that lamivudine does not directly inhibit EBOV RNA polymerase or replication of the virus. Systemic and off-target effects, while not previously described, might be possible. To address this possibility, we plan to assess lamivudine in the mouse model of EVD and will report these findings when available. However, on the basis of these in vitro tests, there is no foundation for recommending lamivudine for treatment of EVD in human patients.


PLOS Neglected Tropical Diseases | 2017

The phosphatidylinositol-3-phosphate 5-kinase inhibitor apilimod blocks filoviral entry and infection

Elizabeth A. Nelson; Julie Dyall; Thomas Hoenen; Alyson B. Barnes; Huanying Zhou; Janie Y. Liang; Julia Michelotti; William H. Dewey; Lisa Evans DeWald; Richard S. Bennett; Patrick J. Morris; Rajarshi Guha; Carleen Klumpp-Thomas; Crystal McKnight; Yu-Chi Chen; Xin Xu; Amy Wang; Emma Hughes; Scott E. Martin; Craig J. Thomas; Peter B. Jahrling; Lisa E. Hensley; Gene G. Olinger; Judith M. White

Phosphatidylinositol-3-phosphate 5-kinase (PIKfyve) is a lipid kinase involved in endosome maturation that emerged from a haploid genetic screen as being required for Ebola virus (EBOV) infection. Here we analyzed the effects of apilimod, a PIKfyve inhibitor that was reported to be well tolerated in humans in phase 2 clinical trials, for its effects on entry and infection of EBOV and Marburg virus (MARV). We first found that apilimod blocks infections by EBOV and MARV in Huh 7, Vero E6 and primary human macrophage cells, with notable potency in the macrophages (IC50, 10 nM). We next observed that similar doses of apilimod block EBOV-glycoprotein-virus like particle (VLP) entry and transcription-replication competent VLP infection, suggesting that the primary mode of action of apilimod is as an entry inhibitor, preventing release of the viral genome into the cytoplasm to initiate replication. After providing evidence that the anti-EBOV action of apilimod is via PIKfyve, we showed that it blocks trafficking of EBOV VLPs to endolysosomes containing Niemann-Pick C1 (NPC1), the intracellular receptor for EBOV. Concurrently apilimod caused VLPs to accumulate in early endosome antigen 1-positive endosomes. We did not detect any effects of apilimod on bulk endosome acidification, on the activity of cathepsins B and L, or on cholesterol export from endolysosomes. Hence by antagonizing PIKfyve, apilimod appears to block EBOV trafficking to its site of fusion and entry into the cytoplasm. Given the drug’s observed anti-filoviral activity, relatively unexplored mechanism of entry inhibition, and reported tolerability in humans, we propose that apilimod be further explored as part of a therapeutic regimen to treat filoviral infections.


PLOS ONE | 2016

Evaluation of the Activity of Lamivudine and Zidovudine against Ebola Virus

Yu Cong; Julie Dyall; Brit J. Hart; Lisa Evans DeWald; Joshua C. Johnson; Elena Postnikova; Huanying Zhou; Robin Gross; Oscar Rojas; Isis Alexander; Nicole Josleyn; Tengfei Zhang; Julia Michelotti; Krisztina Janosko; Pamela J. Glass; Mike Flint; Laura K. McMullan; Christina F. Spiropoulou; Tim Mierzwa; Rajarshi Guha; Paul Shinn; Sam Michael; Carleen Klumpp-Thomas; Crystal McKnight; Craig J. Thomas; Ann E. Eakin; Kathleen O’Loughlin; Carol E. Green; Paul Catz; Jon C. Mirsalis

In the fall of 2014, an international news agency reported that patients suffering from Ebola virus disease (EVD) in Liberia were treated successfully with lamivudine, an antiviral drug used to treat human immunodeficiency virus-1 and hepatitis B virus infections. According to the report, 13 out of 15 patients treated with lamivudine survived and were declared free from Ebola virus disease. In this study, the anti-Ebola virus (EBOV) activity of lamivudine and another antiretroviral, zidovudine, were evaluated in a diverse set of cell lines against two variants of wild-type EBOV. Variable assay parameters were assessed to include different multiplicities of infection, lengths of inoculation times, and durations of dosing. At a multiplicity of infection of 1, lamivudine and zidovudine had no effect on EBOV propagation in Vero E6, Hep G2, or HeLa cells, or in primary human monocyte-derived macrophages. At a multiplicity of infection of 0.1, zidovudine demonstrated limited anti-EBOV activity in Huh 7 cells. Under certain conditions, lamivudine had low anti-EBOV activity at the maximum concentration tested (320 μM). However, lamivudine never achieved greater than 30% viral inhibition, and the activity was not consistently reproducible. Combination of lamivudine and zidovudine showed no synergistic antiviral activity. Independently, a set of in vitro experiments testing lamivudine and zidovudine for antiviral activity against an Ebola-enhanced green fluorescent protein reporter virus was performed at the Centers for Disease Control and Prevention. No antiviral activity was observed for either compound. A study evaluating the efficacy of lamivudine in a guinea pig model of EVD found no survival benefit. This lack of benefit was observed despite plasma lamivudine concentrations in guinea pig of about 4 μg/ml obtained in a separately conducted pharmacokinetics study. These studies found no evidence to support the therapeutic use of lamivudine for the treatment of EVD.


PLOS ONE | 2013

Poxvirus Antigen Staining of Immune Cells as a Biomarker to Predict Disease Outcome in Monkeypox and Cowpox Virus Infection in Non-Human Primates

Haifeng Song; Krisztina Janosko; Reed F. Johnson; Jing Qin; Nicole Josleyn; Catherine Jett; Russell Byrum; Marisa St. Claire; Julie Dyall; Joseph E. Blaney; Gerald Jennings; Peter B. Jahrling

Infection of non-human primates (NHPs) such as rhesus and cynomolgus macaques with monkeypox virus (MPXV) or cowpox virus (CPXV) serve as models to study poxvirus pathogenesis and to evaluate vaccines and anti-orthopox therapeutics. Intravenous inoculation of macaques with high dose of MPXV (>1–2×107 PFU) or CPXV (>102 PFU) results in 80% to 100% mortality and 66 to 100% mortality respectively. Here we report that NHPs with positive detection of poxvirus antigens in immune cells by flow cytometric staining, especially in monocytes and granulocytes succumbed to virus infection and that early positive pox staining is a strong predictor for lethality. Samples from four independent studies were analyzed. Eighteen NHPs from three different experiments were inoculated with two different MPXV strains at lethal doses. Ten NHPs displayed positive pox-staining and all 10 NHPs reached moribund endpoint. In contrast, none of the three NHPs that survived anticipated lethal virus dose showed apparent virus staining in the monocytes and granulocytes. In addition, three NHPs that were challenged with a lethal dose of MPXV and received cidofovir treatment were pox-antigen negative and all three NHPs survived. Furthermore, data from a CPXV study also demonstrated that 6/9 NHPs were pox-antigen staining positive and all 6 NHPs reached euthanasia endpoint, while the three survivors were pox-antigen staining negative. Thus, we conclude that monitoring pox-antigen staining in immune cells can be used as a biomarker to predict the prognosis of virus infection. Future studies should focus on the mechanisms and implications of the pox-infection of immune cells and the correlation between pox-antigen detection in immune cells and disease progression in human poxviral infection.

Collaboration


Dive into the Julie Dyall's collaboration.

Top Co-Authors

Avatar

Peter B. Jahrling

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Lisa E. Hensley

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Gene G. Olinger

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Reed F. Johnson

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Brit J. Hart

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Huanying Zhou

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Elena Postnikova

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Joseph E. Blaney

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Richard S. Bennett

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge