Network


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

Hotspot


Dive into the research topics where Shannan L. Rossi is active.

Publication


Featured researches published by Shannan L. Rossi.


American Journal of Tropical Medicine and Hygiene | 2016

Characterization of a Novel Murine Model to Study Zika Virus

Shannan L. Rossi; Robert B. Tesh; Sasha R. Azar; Antonio E. Muruato; Kathryn A. Hanley; Albert J. Auguste; Rose M. Langsjoen; Slobodan Paessler; Nikos Vasilakis; Scott C. Weaver

The mosquito-borne Zika virus (ZIKV) is responsible for an explosive ongoing outbreak of febrile illness across the Americas. ZIKV was previously thought to cause only a mild, flu-like illness, but during the current outbreak, an association with Guillain–Barré syndrome and microcephaly in neonates has been detected. A previous study showed that ZIKV requires murine adaptation to generate reproducible murine disease. In our study, a low-passage Cambodian isolate caused disease and mortality in mice lacking the interferon (IFN) alpha receptor (A129 mice) in an age-dependent manner, but not in similarly aged immunocompetent mice. In A129 mice, viremia peaked at ∼107 plaque-forming units/mL by day 2 postinfection (PI) and reached high titers in the spleen by day 1. ZIKV was detected in the brain on day 3 PI and caused signs of neurologic disease, including tremors, by day 6. Robust replication was also noted in the testis. In this model, all mice infected at the youngest age (3 weeks) succumbed to illness by day 7 PI. Older mice (11 weeks) showed signs of illness, viremia, and weight loss but recovered starting on day 8. In addition, AG129 mice, which lack both type I and II IFN responses, supported similar infection kinetics to A129 mice, but with exaggerated disease signs. This characterization of an Asian lineage ZIKV strain in a murine model, and one of the few studies reporting a model of Zika disease and demonstrating age-dependent morbidity and mortality, could provide a platform for testing the efficacy of antivirals and vaccines.


Nature Communications | 2014

Multi-peaked adaptive landscape for chikungunya virus evolution predicts continued fitness optimization in Aedes albopictus mosquitoes

Konstantin A. Tsetsarkin; Rubing Chen; Ruimei Yun; Shannan L. Rossi; Kenneth Plante; Mathilde Guerbois; Naomi L. Forrester; Guey Chuen Perng; Easwaran Sreekumar; Grace Leal; Jing Huang; Suchetana Mukhopadhyay; Scott C. Weaver

Host species-specific fitness landscapes largely determine the outcome of host switching during pathogen emergence. Using chikungunya virus (CHIKV) to study adaptation to a mosquito vector, we evaluated mutations associated with recently evolved sub-lineages. Multiple Aedes albopictus-adaptive fitness peaks became available after CHIKV acquired an initial adaptive (E1-A226V) substitution, permitting rapid lineage diversification observed in nature. All second-step mutations involved replacements by glutamine or glutamic acid of E2 glycoprotein amino acids in the acid-sensitive region, providing a framework to anticipate additional A. albopictus-adaptive mutations. The combination of second-step adaptive mutations into a single, ‘super-adaptive’ fitness peak also predicted the future emergence of CHIKV strains with even greater transmission efficiency in some current regions of endemic circulation, followed by their likely global spread. Supplementary information The online version of this article (doi:10.1038/ncomms5084) contains supplementary material, which is available to authorized users.


PLOS Pathogens | 2009

Mosquitoes Put the Brake on Arbovirus Evolution: Experimental Evolution Reveals Slower Mutation Accumulation in Mosquito Than Vertebrate Cells

Nikos Vasilakis; Eleanor R. Deardorff; Joan L. Kenney; Shannan L. Rossi; Kathryn A. Hanley; Scott C. Weaver

Like other arthropod-borne viruses (arboviruses), mosquito-borne dengue virus (DENV) is maintained in an alternating cycle of replication in arthropod and vertebrate hosts. The trade-off hypothesis suggests that this alternation constrains DENV evolution because a fitness increase in one host usually diminishes fitness in the other. Moreover, the hypothesis predicts that releasing DENV from host alternation should facilitate adaptation. To test this prediction, DENV was serially passaged in either a single human cell line (Huh-7), a single mosquito cell line (C6/36), or in alternating passages between Huh-7 and C6/36 cells. After 10 passages, consensus mutations were identified and fitness was assayed by evaluating replication kinetics in both cell types as well as in a novel cell type (Vero) that was not utilized in any of the passage series. Viruses allowed to specialize in single host cell types exhibited fitness gains in the cell type in which they were passaged, but fitness losses in the bypassed cell type, and most alternating passages, exhibited fitness gains in both cell types. Interestingly, fitness gains were observed in the alternately passaged, cloned viruses, an observation that may be attributed to the acquisition of both host cell–specific and amphi-cell-specific adaptations or to recovery from the fitness losses due to the genetic bottleneck of biological cloning. Amino acid changes common to both passage series suggested convergent evolution to replication in cell culture via positive selection. However, intriguingly, mutations accumulated more rapidly in viruses passed in Huh-7 cells than in those passed in C6/36 cells or in alternation. These results support the hypothesis that releasing DENV from host alternation facilitates adaptation, but there is limited support for the hypothesis that such alternation necessitates a fitness trade-off. Moreover, these findings suggest that patterns of genetic evolution may differ between viruses replicating in mammalian and mosquito cells.


Nature Medicine | 2017

A live-attenuated Zika virus vaccine candidate induces sterilizing immunity in mouse models

Chao Shan; Antonio E. Muruato; Bruno T.D. Nunes; Huanle Luo; Xuping Xie; Daniele Barbosa de Almeida Medeiros; Maki Wakamiya; Robert B. Tesh; Alan D. T. Barrett; Tian Wang; Scott C. Weaver; Pedro Fernando da Costa Vasconcelos; Shannan L. Rossi; Pei Yong Shi

Zika virus (ZIKV) infection of pregnant women can cause a wide range of congenital abnormalities, including microcephaly, in the infant, a condition now collectively known as congenital ZIKV syndrome. A vaccine to prevent or significantly attenuate viremia in pregnant women who are residents of or travelers to epidemic or endemic regions is needed to avert congenital ZIKV syndrome, and might also help to suppress epidemic transmission. Here we report on a live-attenuated vaccine candidate that contains a 10-nucleotide deletion in the 3′ untranslated region of the ZIKV genome (10-del ZIKV). The 10-del ZIKV is highly attenuated, immunogenic, and protective in type 1 interferon receptor–deficient A129 mice. Crucially, a single dose of 10-del ZIKV induced sterilizing immunity with a saturated neutralizing antibody titer, which no longer increased after challenge with an epidemic ZIKV, and completely prevented viremia. The immunized mice also developed a robust T cell response. Intracranial inoculation of 1-d-old immunocompetent CD-1 mice with 1 × 104 infectious focus units (IFU) of 10-del ZIKV caused no mortality, whereas infections with 10 IFU of wild-type ZIKV were lethal. Mechanistically, the attenuated virulence of 10-del ZIKV may be due to decreased viral RNA synthesis and increased sensitivity to type-1-interferon inhibition. The attenuated 10-del ZIKV was incapable of infecting mosquitoes after oral feeding of spiked-blood meals, representing an additional safety feature. Collectively, the safety and efficacy results suggest that further development of this promising, live-attenuated ZIKV vaccine candidate is warranted.


Journal of Virology | 2013

Negevirus: a Proposed New Taxon of Insect-Specific Viruses with Wide Geographic Distribution

Nikos Vasilakis; Naomi L. Forrester; Gustavo Palacios; Farooq Nasar; Nazir Savji; Shannan L. Rossi; Hilda Guzman; Thomas G. Wood; Vsevolod L. Popov; Rodion Gorchakov; Ana Vázquez González; Andrew D. Haddow; Douglas M. Watts; Amelia Travassos da Rosa; Scott C. Weaver; W. Ian Lipkin; Robert B. Tesh

ABSTRACT Six novel insect-specific viruses, isolated from mosquitoes and phlebotomine sand flies collected in Brazil, Peru, the United States, Ivory Coast, Israel, and Indonesia, are described. Their genomes consist of single-stranded, positive-sense RNAs with poly(A) tails. By electron microscopy, the virions appear as spherical particles with diameters of ∼45 to 55 nm. Based on their genome organization and phylogenetic relationship, the six viruses, designated Negev, Ngewotan, Piura, Loreto, Dezidougou, and Santana, appear to form a new taxon, tentatively designated Negevirus. Their closest but still distant relatives are citrus leposis virus C (CiLV-C) and viruses in the genus Cilevirus, which are mite-transmitted plant viruses. The negeviruses replicate rapidly and to high titer (up to 1010 PFU/ml) in mosquito cells, producing extensive cytopathic effect and plaques, but they do not appear to replicate in mammalian cells or mice. A discussion follows on their possible biological significance and effect on mosquito vector competence for arboviruses.


Journal of Virology | 2012

Attenuation of Chikungunya Virus Vaccine Strain 181/Clone 25 Is Determined by Two Amino Acid Substitutions in the E2 Envelope Glycoprotein

Rodion Gorchakov; Eryu Wang; Grace Leal; Naomi L. Forrester; Kenneth Plante; Shannan L. Rossi; Charalambos D. Partidos; A. P. Adams; Robert L. Seymour; James Weger; Erin M. Borland; Michael B. Sherman; Ann M. Powers; Jorge E. Osorio; Scott C. Weaver

ABSTRACT Chikungunya virus (CHIKV) is the mosquito-borne alphavirus that is the etiologic agent of massive outbreaks of arthralgic febrile illness that recently affected millions of people in Africa and Asia. The only CHIKV vaccine that has been tested in humans, strain 181/clone 25, is a live-attenuated derivative of Southeast Asian human isolate strain AF15561. The vaccine was immunogenic in phase I and II clinical trials; however, it induced transient arthralgia in 8% of the vaccinees. There are five amino acid differences between the vaccine and its parent, as well as five synonymous mutations, none of which involves cis-acting genome regions known to be responsible for replication or packaging. To identify the determinants of attenuation, we therefore tested the five nonsynonymous mutations by cloning them individually or in different combinations into infectious clones derived from two wild-type (WT) CHIKV strains, La Reunion and AF15561. Levels of virulence were compared with those of the WT strains and the vaccine strain in two different murine models: infant CD1 and adult A129 mice. An attenuated phenotype indistinguishable from that of the 181/clone 25 vaccine strain was obtained by the simultaneous expression of two E2 glycoprotein substitutions, with intermediate levels of attenuation obtained with the single E2 mutations. The other three amino acid mutations, in nsP1, 6K, and E1, did not have a detectable effect on CHIKV virulence. These results indicate that the attenuation of strain 181/clone 25 is mediated by two point mutations, explaining the phenotypic instability observed in human vaccinees and also in our studies.


Cell | 2017

Vaccine Mediated Protection Against Zika Virus-Induced Congenital Disease

Justin M. Richner; Brett W. Jagger; Chao Shan; Camila R. Fontes; Kimberly A. Dowd; Bin Cao; Sunny Himansu; Elizabeth A. Caine; Bruno T.D. Nunes; Daniele Barbosa de Almeida Medeiros; Antonio E. Muruato; Bryant M. Foreman; Huanle Luo; Tian Wang; Alan D. T. Barrett; Scott C. Weaver; Pedro Fernando da Costa Vasconcelos; Shannan L. Rossi; Giuseppe Ciaramella; Indira U. Mysorekar; Theodore C. Pierson; Pei Yong Shi; Michael S. Diamond

The emergence of Zika virus (ZIKV) and its association with congenital malformations has prompted the rapid development of vaccines. Although efficacy with multiple viral vaccine platforms has been established in animals, no study has addressed protection during pregnancy. We tested in mice two vaccine platforms, a lipid nanoparticle-encapsulated modified mRNA vaccine encoding ZIKV prM and E genes and a live-attenuated ZIKV strain encoding an NS1 protein without glycosylation, for their ability to protect against transmission to the fetus. Vaccinated dams challenged with a heterologous ZIKV strain at embryo day 6 (E6) and evaluated at E13 showed markedly diminished levels of viral RNA in maternal, placental, and fetal tissues, which resulted in protection against placental damage and fetal demise. As modified mRNA and live-attenuated vaccine platforms can restrict in utero transmission of ZIKV in mice, their further development in humans to prevent congenital ZIKV syndrome is warranted.


Journal of Virology | 2007

Early Production of Type I Interferon during West Nile Virus Infection: Role for Lymphoid Tissues in IRF3-Independent Interferon Production

Nigel Bourne; Frank Scholle; Maria Carlan Silva; Shannan L. Rossi; Nathan Dewsbury; Barbara M. Judy; Juliana B. de Aguiar; Megan A. Leon; D. Mark Estes; Rafik Fayzulin; Peter W. Mason

ABSTRACT Infection of cells with flaviviruses in vitro is reduced by pretreatment with small amounts of type I interferon (IFN-α/β). Similarly, pretreatment of animals with IFN and experiments using mice defective in IFN signaling have indicated a role for IFN in controlling flavivirus disease in vivo. These data, along with findings that flavivirus-infected cells block IFN signaling, suggest that flavivirus infection can trigger an IFN response. To investigate IFN gene induction by the very first cells infected during in vivo infection with the flavivirus West Nile virus (WNV), we infected mice with high-titer preparations of WNV virus-like particles (VLPs), which initiate viral genome replication in cells but fail to spread. These studies demonstrated a brisk production of IFN in vivo, with peak levels of over 1,000 units/ml detected in sera between 8 and 24 h after inoculation by either the intraperitoneal or footpad route. The IFN response was dependent on genome replication, and WNV genomes and WNV antigen-positive cells were readily detected in the popliteal lymph nodes (pLN) of VLP-inoculated mice. High levels of IFN mRNA transcripts and functional IFN were also produced in VLP-inoculated IFN regulatory factor 3 null (IRF3−/−) mice, indicating that IFN production was independent of the IRF3 pathways to IFN gene transcription, consistent with the IFN type produced (predominantly α).


Infection, Genetics and Evolution | 2013

Fever versus fever: The role of host and vector susceptibility and interspecific competition in shaping the current and future distributions of the sylvatic cycles of dengue virus and yellow fever virus

Kathryn A. Hanley; Thomas P. Monath; Scott C. Weaver; Shannan L. Rossi; Rebecca Richman; Nikos Vasilakis

Two different species of flaviviruses, dengue virus (DENV) and yellow fever virus (YFV), that originated in sylvatic cycles maintained in non-human primates and forest-dwelling mosquitoes have emerged repeatedly into sustained human-to-human transmission by Aedes aegypti mosquitoes. Sylvatic cycles of both viruses remain active, and where the two viruses overlap in West Africa they utilize similar suites of monkeys and Aedes mosquitoes. These extensive similarities render the differences in the biogeography and epidemiology of the two viruses all the more striking. First, the sylvatic cycle of YFV originated in Africa and was introduced into the New World, probably as a result of the slave trade, but is absent in Asia; in contrast, sylvatic DENV likely originated in Asia and has spread to Africa but not to the New World. Second, while sylvatic YFV can emerge into extensive urban outbreaks in humans, these invariably die out, whereas four different types of DENV have established human transmission cycles that are ecologically and evolutionarily distinct from their sylvatic ancestors. Finally, transmission of YFV among humans has been documented only in Africa and the Americas, whereas DENV is transmitted among humans across most of the range of competent Aedes vectors, which in the last decade has included every continent save Antarctica. This review summarizes current understanding of sylvatic transmission cycles of YFV and DENV, considers possible explanations for their disjunct distributions, and speculates on the potential consequences of future establishment of a sylvatic cycle of DENV in the Americas.


Emerging Infectious Diseases | 2017

Variation in aedes aegypti mosquito competence for zika virus transmission

Christopher M. Roundy; Sasha R. Azar; Shannan L. Rossi; Jing H. Huang; Grace Leal; Ruimei Yun; Ildefonso Fernández-Salas; Christopher J. Vitek; Igor Adolfo Dexheimer Paploski; Uriel Kitron; Guilherme S. Ribeiro; Kathryn A. Hanley; Scott C. Weaver; Nikos Vasilakis

To test whether Zika virus has adapted for more efficient transmission by Aedes aegypti mosquitoes, leading to recent urban outbreaks, we fed mosquitoes from Brazil, the Dominican Republic, and the United States artificial blood meals containing 1 of 3 Zika virus strains (Senegal, Cambodia, Mexico) and monitored infection, dissemination, and virus in saliva. Contrary to our hypothesis, Cambodia and Mexica strains were less infectious than the Senegal strain. Only mosquitoes from the Dominican Republic transmitted the Cambodia and Mexica strains. However, blood meals from viremic mice were more infectious than artificial blood meals of comparable doses; the Cambodia strain was not transmitted by mosquitoes from Brazil after artificial blood meals, whereas 61% transmission occurred after a murine blood meal (saliva titers up to 4 log10 infectious units/collection). Although regional origins of vector populations and virus strain influence transmission efficiency, Ae. aegypti mosquitoes appear to be competent vectors of Zika virus in several regions of the Americas.

Collaboration


Dive into the Shannan L. Rossi's collaboration.

Top Co-Authors

Avatar

Scott C. Weaver

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Nikos Vasilakis

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Chao Shan

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Christopher M. Roundy

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Pei Yong Shi

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Sasha R. Azar

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Antonio E. Muruato

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Grace Leal

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Xuping Xie

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Naomi L. Forrester

University of Texas Medical Branch

View shared research outputs
Researchain Logo
Decentralizing Knowledge