Network


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

Hotspot


Dive into the research topics where Elyse R. Nagle is active.

Publication


Featured researches published by Elyse R. Nagle.


Nature | 2016

Therapeutic efficacy of the small molecule GS-5734 against Ebola virus in rhesus monkeys

Travis K. Warren; Robert Jordan; Michael K. Lo; Adrian S. Ray; Richard L. Mackman; Veronica Soloveva; Dustin Siegel; Michel Perron; Roy Bannister; Hon C. Hui; Nate Larson; Robert Strickley; Jay Wells; Kelly S. Stuthman; Sean A. Van Tongeren; Nicole L. Garza; Ginger Donnelly; Amy C. Shurtleff; Cary J. Retterer; Dima Gharaibeh; Rouzbeh Zamani; Tara Kenny; Brett P. Eaton; Elizabeth Grimes; Lisa S. Welch; Laura Gomba; Catherine L. Wilhelmsen; Donald K. Nichols; Jonathan E. Nuss; Elyse R. Nagle

The most recent Ebola virus outbreak in West Africa, which was unprecedented in the number of cases and fatalities, geographic distribution, and number of nations affected, highlights the need for safe, effective, and readily available antiviral agents for treatment and prevention of acute Ebola virus (EBOV) disease (EVD) or sequelae. No antiviral therapeutics have yet received regulatory approval or demonstrated clinical efficacy. Here we report the discovery of a novel small molecule GS-5734, a monophosphoramidate prodrug of an adenosine analogue, with antiviral activity against EBOV. GS-5734 exhibits antiviral activity against multiple variants of EBOV and other filoviruses in cell-based assays. The pharmacologically active nucleoside triphosphate (NTP) is efficiently formed in multiple human cell types incubated with GS-5734 in vitro, and the NTP acts as an alternative substrate and RNA-chain terminator in primer-extension assays using a surrogate respiratory syncytial virus RNA polymerase. Intravenous administration of GS-5734 to nonhuman primates resulted in persistent NTP levels in peripheral blood mononuclear cells (half-life, 14 h) and distribution to sanctuary sites for viral replication including testes, eyes, and brain. In a rhesus monkey model of EVD, once-daily intravenous administration of 10 mg kg−1 GS-5734 for 12 days resulted in profound suppression of EBOV replication and protected 100% of EBOV-infected animals against lethal disease, ameliorating clinical disease signs and pathophysiological markers, even when treatments were initiated three days after virus exposure when systemic viral RNA was detected in two out of six treated animals. These results show the first substantive post-exposure protection by a small-molecule antiviral compound against EBOV in nonhuman primates. The broad-spectrum antiviral activity of GS-5734 in vitro against other pathogenic RNA viruses, including filoviruses, arenaviruses, and coronaviruses, suggests the potential for wider medical use. GS-5734 is amenable to large-scale manufacturing, and clinical studies investigating the drug safety and pharmacokinetics are ongoing.


Nature | 2017

Genomic epidemiology reveals multiple introductions of Zika virus into the United States

Nathan D. Grubaugh; Jason T. Ladner; Moritz U. G. Kraemer; Gytis Dudas; Amanda L. Tan; Karthik Gangavarapu; Michael R. Wiley; Stephen White; Julien Thézé; Diogo M. Magnani; Karla Prieto; Daniel Reyes; Andrea M. Bingham; Lauren M. Paul; Refugio Robles-Sikisaka; Glenn Oliveira; Darryl Pronty; Carolyn M. Barcellona; Hayden C. Metsky; Mary Lynn Baniecki; Kayla G. Barnes; Bridget Chak; Catherine A. Freije; Adrianne Gladden-Young; Andreas Gnirke; Cynthia Y. Luo; Bronwyn MacInnis; Christian B. Matranga; Daniel J. Park; James Qu

Zika virus (ZIKV) is causing an unprecedented epidemic linked to severe congenital abnormalities. In July 2016, mosquito-borne ZIKV transmission was reported in the continental United States; since then, hundreds of locally acquired infections have been reported in Florida. To gain insights into the timing, source, and likely route(s) of ZIKV introduction, we tracked the virus from its first detection in Florida by sequencing ZIKV genomes from infected patients and Aedes aegypti mosquitoes. We show that at least 4 introductions, but potentially as many as 40, contributed to the outbreak in Florida and that local transmission is likely to have started in the spring of 2016—several months before its initial detection. By analysing surveillance and genetic data, we show that ZIKV moved among transmission zones in Miami. Our analyses show that most introductions were linked to the Caribbean, a finding corroborated by the high incidence rates and traffic volumes from the region into the Miami area. Our study provides an understanding of how ZIKV initiates transmission in new regions.


Cell Host & Microbe | 2015

Evolution and Spread of Ebola Virus in Liberia, 2014–2015

Jason T. Ladner; Michael R. Wiley; Suzanne Mate; Gytis Dudas; Karla Prieto; Sean Lovett; Elyse R. Nagle; Brett Beitzel; Merle L. Gilbert; Lawrence S. Fakoli; Joseph W. Diclaro; Randal J. Schoepp; Joseph N. Fair; Jens H. Kuhn; Lisa E. Hensley; Daniel J. Park; Pardis C. Sabeti; Andrew Rambaut; Mariano Sanchez-Lockhart; Fatorma K. Bolay; Jeffrey R. Kugelman; Gustavo Palacios

The 2013-present Western African Ebola virus disease (EVD) outbreak is the largest ever recorded with >28,000 reported cases. Ebola virus (EBOV) genome sequencing has played an important role throughout this outbreak; however, relatively few sequences have been determined from patients in Liberia, the second worst-affected country. Here, we report 140 EBOV genome sequences from the second wave of the Liberian outbreak and analyze them in combination with 782 previously published sequences from throughout the Western African outbreak. While multiple early introductions of EBOV to Liberia are evident, the majority of Liberian EVD cases are consistent with a single introduction, followed by spread and diversification within the country. Movement of the virus within Liberia was widespread, and reintroductions from Liberia served as an important source for the continuation of the already ongoing EVD outbreak in Guinea. Overall, little evidence was found for incremental adaptation of EBOV to the human host.


Cell Reports | 2015

Emergence of Ebola Virus Escape Variants in Infected Nonhuman Primates Treated with the MB-003 Antibody Cocktail.

Jeffrey R. Kugelman; Johanny Kugelman-Tonos; Jason T. Ladner; James Pettit; Carolyn M. Keeton; Elyse R. Nagle; Karla Garcia; Jeffrey W. Froude; Ana I. Kuehne; Jens H. Kuhn; Sina Bavari; Larry Zeitlin; John M. Dye; Gene G. Olinger; Mariano Sanchez-Lockhart; Gustavo Palacios

MB-003, a plant-derived monoclonal antibody cocktail used effectively in treatment of Ebola virus infection in non-human primates, was unable to protect two of six animals when initiated 1 or 2 days post-infection. We characterized a mechanism of viral escape in one of the animals, after observation of two clusters of genomic mutations that resulted in five nonsynonymous mutations in the monoclonal antibody target sites. These mutations were linked to a reduction in antibody binding and later confirmed to be present in a viral isolate that was not neutralized in vitro. Retrospective evaluation of a second independent study allowed the identification of a similar case. Four SNPs in previously identified positions were found in this second fatality, suggesting that genetic drift could be a potential cause for treatment failure. These findings highlight the importance selecting different target domains for each component of the cocktail to minimize the potential for viral escape.


Genome Announcements | 2016

Complete Genome Sequences of Five Zika Virus Isolates

Jason T. Ladner; Michael R. Wiley; Karla Prieto; Chadwick Yasuda; Elyse R. Nagle; Matthew R. Kasper; Daniel Reyes; Nikolaos Vasilakis; Vireak Heang; Scott C. Weaver; Andrew D. Haddow; Robert B. Tesh; Ly Sovann; Gustavo Palacios

ABSTRACT Zika virus is an emerging human pathogen of great concern due to putative links to microcephaly and Guillain-Barre syndrome. Here, we report the complete genomes, including the 5′ and 3′ untranslated regions, of five Zika virus isolates, one from the Asian lineage and four from the African lineage.


BMC Genomics | 2014

Viral diversity and clonal evolution from unphased genomic data

Hossein Khiabanian; Zachary Carpenter; Jeffrey R. Kugelman; Joseph Chan; Vladimir Trifonov; Elyse R. Nagle; Travis K. Warren; Patrick L. Iversen; Sina Bavari; Gustavo Palacios; Raul Rabadan

BackgroundClonal expansion is a process in which a single organism reproduces asexually, giving rise to a diversifying population. It is pervasive in nature, from within-host pathogen evolution to emergent infectious disease outbreaks. Standard phylogenetic tools rely on full-length genomes of individual pathogens or population consensus sequences (phased genotypes).Although high-throughput sequencing technologies are able to sample population diversity, the short sequence reads inherent to them preclude assessing whether two reads originate from the same clone (unphased genotypes). This obstacle severely limits the application of phylogenetic methods and investigation of within-host dynamics of acute infections using this rich data source.MethodsWe introduce two measures of diversity to study the evolution of clonal populations using unphased genomic data, which eliminate the need to construct full-length genomes. Our method follows a maximum likelihood approach to estimate evolutionary rates and times to the most recent common ancestor, based on a relaxed molecular clock model; independent of a growth model. Deviations from neutral evolution indicate the presence of selection and bottleneck events.ResultsWe evaluated our methods in silico and then compared it against existing approaches with the well-characterized 2009 H1N1 influenza pandemic. We then applied our method to high-throughput genomic data from marburgvirus-infected non-human primates and inferred the time of infection and the intra-host evolutionary rate, and identified purifying selection in viral populations.ConclusionsOur method has the power to make use of minor variants present in less than 1% of the population and capture genomic diversification within days of infection, making it an ideal tool for the study of acute RNA viral infection dynamics.


BMC Genomics | 2015

De novo transcriptome reconstruction and annotation of the Egyptian rousette bat

Albert Lee; Kirsten Kulcsar; Oliver Elliott; Hossein Khiabanian; Elyse R. Nagle; Megan E. B. Jones; Brian R. Amman; Mariano Sanchez-Lockhart; Jonathan S. Towner; Gustavo Palacios; Raul Rabadan

BackgroundThe Egyptian Rousette bat (Rousettus aegyptiacus), a common fruit bat species found throughout Africa and the Middle East, was recently identified as a natural reservoir host of Marburg virus. With Ebola virus, Marburg virus is a member of the family Filoviridae that causes severe hemorrhagic fever disease in humans and nonhuman primates, but results in little to no pathological consequences in bats. Understanding host-pathogen interactions within reservoir host species and how it differs from hosts that experience severe disease is an important aspect of evaluating viral pathogenesis and developing novel therapeutics and methods of prevention.ResultsProgress in studying bat reservoir host responses to virus infection is hampered by the lack of host-specific reagents required for immunological studies. In order to establish a basis for the design of reagents, we sequenced, assembled, and annotated the R. aegyptiacus transcriptome. We performed de novo transcriptome assembly using deep RNA sequencing data from 11 distinct tissues from one male and one female bat. We observed high similarity between this transcriptome and those available from other bat species. Gene expression analysis demonstrated clustering of expression profiles by tissue, where we also identified enrichment of tissue-specific gene ontology terms. In addition, we identified and experimentally validated the expression of novel coding transcripts that may be specific to this species.ConclusionWe comprehensively characterized the R. aegyptiacus transcriptome de novo. This transcriptome will be an important resource for understanding bat immunology, physiology, disease pathogenesis, and virus transmission.


BMC Genomics | 2014

Transcriptome reconstruction and annotation of cynomolgus and African green monkey

Albert Lee; Hossein Khiabanian; Jeffrey R. Kugelman; Oliver Elliott; Elyse R. Nagle; Guo-Yun Yu; Travis K. Warren; Gustavo Palacios; Raul Rabadan

BackgroundNon-human primates (NHPs) and humans share major biological mechanisms, functions, and responses due to their close evolutionary relationship and, as such, provide ideal animal models to study human diseases. RNA expression in NHPs provides specific signatures that are informative of disease mechanisms and therapeutic modes of action. Unlike the human transcriptome, the transcriptomes of major NHP animal models are yet to be comprehensively annotated.ResultsIn this manuscript, employing deep RNA sequencing of seven tissue samples, we characterize the transcriptomes of two commonly used NHP animal models: Cynomolgus macaque (Macaca fascicularis) and African green monkey (Chlorocebus aethiops). We present the Multi-Species Annotation (MSA) pipeline that leverages well-annotated primate species and annotates 99.8% of reconstructed transcripts. We elucidate tissue-specific expression profiles and report 13 experimentally validated novel transcripts in these NHP animal models.ConclusionWe report comprehensively annotated transcriptomes of two non-human primates, which we have made publically available on a customized UCSC Genome Browser interface. The MSA pipeline is also freely available.


Scientific Reports | 2017

Comparison of Transcriptomic Platforms for Analysis of Whole Blood from Ebola-Infected Cynomolgus Macaques

Emily Speranza; Louis A. Altamura; Kirsten Kulcsar; Sandra L. Bixler; Cynthia A. Rossi; Randal J. Schoepp; Elyse R. Nagle; William Aguilar; Christina E. Douglas; Korey L. Delp; Timothy D. Minogue; Gustavo Palacios; Arthur J. Goff; John H. Connor

Ebola virus disease (EVD) is a serious illness with mortality rates of 20–90% in various outbreaks. EVD is characterized by robust virus replication and strong host inflammatory response. Analyzing host immune responses has increasingly involved multimodal approaches including transcriptomics to profile gene expression. We studied cynomolgus macaques exposed to Ebola virus Makona via different routes with the intent of comparing RNA-Seq to a NanoString nCounter codeset targeting 769 non-human primate (NHP) genes. RNA-Seq analysis of serial blood samples showed different routes led to the same overall transcriptional response seen in previously reported EBOV-exposed NHP studies. Both platforms displayed a strong correlation in gene expression patterns, including a strong induction of innate immune response genes at early times post-exposure, and neutrophil-associated genes at later time points. A 41-gene classifier was tested in both platforms for ability to cluster samples by infection status. Both NanoString and RNA-Seq could be used to predict relative abundances of circulating immune cell populations that matched traditional hematology. This demonstrates the complementarity of RNA-Seq and NanoString. Moreover, the development of an NHP-specific NanoString codeset should augment studies of filoviruses and other high containment infectious diseases without the infrastructure requirements of RNA-Seq technology.


PLOS ONE | 2017

Error baseline rates of five sample preparation methods used to characterize RNA virus populations

Jeffrey R. Kugelman; Michael R. Wiley; Elyse R. Nagle; Daniel Reyes; Brad P. Pfeffer; Jens H. Kuhn; Mariano Sanchez-Lockhart; Gustavo Palacios

Individual RNA viruses typically occur as populations of genomes that differ slightly from each other due to mutations introduced by the error-prone viral polymerase. Understanding the variability of RNA virus genome populations is critical for understanding virus evolution because individual mutant genomes may gain evolutionary selective advantages and give rise to dominant subpopulations, possibly even leading to the emergence of viruses resistant to medical countermeasures. Reverse transcription of virus genome populations followed by next-generation sequencing is the only available method to characterize variation for RNA viruses. However, both steps may lead to the introduction of artificial mutations, thereby skewing the data. To better understand how such errors are introduced during sample preparation, we determined and compared error baseline rates of five different sample preparation methods by analyzing in vitro transcribed Ebola virus RNA from an artificial plasmid-based system. These methods included: shotgun sequencing from plasmid DNA or in vitro transcribed RNA as a basic “no amplification” method, amplicon sequencing from the plasmid DNA or in vitro transcribed RNA as a “targeted” amplification method, sequence-independent single-primer amplification (SISPA) as a “random” amplification method, rolling circle reverse transcription sequencing (CirSeq) as an advanced “no amplification” method, and Illumina TruSeq RNA Access as a “targeted” enrichment method. The measured error frequencies indicate that RNA Access offers the best tradeoff between sensitivity and sample preparation error (1.4−5) of all compared methods.

Collaboration


Dive into the Elyse R. Nagle's collaboration.

Top Co-Authors

Avatar

Gustavo Palacios

United States Army Medical Research Institute of Infectious Diseases

View shared research outputs
Top Co-Authors

Avatar

Jason T. Ladner

United States Army Medical Research Institute of Infectious Diseases

View shared research outputs
Top Co-Authors

Avatar

Jeffrey R. Kugelman

United States Army Medical Research Institute of Infectious Diseases

View shared research outputs
Top Co-Authors

Avatar

Michael R. Wiley

United States Army Medical Research Institute of Infectious Diseases

View shared research outputs
Top Co-Authors

Avatar

Karla Prieto

United States Army Medical Research Institute of Infectious Diseases

View shared research outputs
Top Co-Authors

Avatar

Mariano Sanchez-Lockhart

United States Army Medical Research Institute of Infectious Diseases

View shared research outputs
Top Co-Authors

Avatar

Daniel Reyes

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jens H. Kuhn

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge