Network


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

Hotspot


Dive into the research topics where Mariel S. Mohns is active.

Publication


Featured researches published by Mariel S. Mohns.


Nature Communications | 2016

A rhesus macaque model of Asian-lineage Zika virus infection

Dawn M. Dudley; Matthew T. Aliota; Emma L. Mohr; Andrea M. Weiler; Gabrielle Lehrer-Brey; Kim L. Weisgrau; Mariel S. Mohns; Meghan E. Breitbach; Mustafa N. Rasheed; Christina M. Newman; Dane D. Gellerup; Louise H. Moncla; Jennifer Post; Nancy Schultz-Darken; Michele L. Schotzko; Jennifer M. Hayes; Josh Eudailey; M. Anthony Moody; Sallie R. Permar; Shelby L. O’Connor; Eva G. Rakasz; Heather A. Simmons; Saverio Capuano; Thaddeus G. Golos; Jorge E. Osorio; Thomas C. Friedrich; David H. O’Connor

Infection with Asian-lineage Zika virus (ZIKV) has been associated with Guillain–Barré syndrome and fetal abnormalities, but the underlying mechanisms remain poorly understood. Animal models of infection are thus urgently needed. Here we show that rhesus macaques are susceptible to infection by an Asian-lineage ZIKV closely related to strains currently circulating in the Americas. Following subcutaneous inoculation, ZIKV RNA is detected in plasma 1 day post infection (d.p.i.) in all animals (N=8, including 2 pregnant animals), and is also present in saliva, urine and cerebrospinal fluid. Non-pregnant and pregnant animals remain viremic for 21 days and for up to at least 57 days, respectively. Neutralizing antibodies are detected by 21 d.p.i. Rechallenge 10 weeks after the initial challenge results in no detectable virus replication, indicating protective immunity against homologous strains. Therefore, Asian-lineage ZIKV infection of rhesus macaques provides a relevant animal model for studying pathogenesis and evaluating potential interventions against human infection, including during pregnancy.


PLOS Pathogens | 2017

Highly efficient maternal-fetal Zika virus transmission in pregnant rhesus macaques

Sydney Nguyen; Kathleen M. Antony; Dawn M. Dudley; Sarah Kohn; Heather A. Simmons; Bryce Wolfe; M. Shahriar Salamat; Leandro B. C. Teixeira; Gregory J. Wiepz; Troy H. Thoong; Matthew T. Aliota; Andrea M. Weiler; Gabrielle L. Barry; Kim L. Weisgrau; Logan J. Vosler; Mariel S. Mohns; Meghan E. Breitbach; Laurel M. Stewart; Mustafa N. Rasheed; Christina M. Newman; Michael E. Graham; Oliver Wieben; Patrick A. Turski; Kevin M. Johnson; Jennifer Post; Jennifer M. Hayes; Nancy Schultz-Darken; Michele L. Schotzko; Josh Eudailey; Sallie R. Permar

Infection with Zika virus (ZIKV) is associated with human congenital fetal anomalies. To model fetal outcomes in nonhuman primates, we administered Asian-lineage ZIKV subcutaneously to four pregnant rhesus macaques. While non-pregnant animals in a previous study contemporary with the current report clear viremia within 10–12 days, maternal viremia was prolonged in 3 of 4 pregnancies. Fetal head growth velocity in the last month of gestation determined by ultrasound assessment of head circumference was decreased in comparison with biparietal diameter and femur length within each fetus, both within normal range. ZIKV RNA was detected in tissues from all four fetuses at term cesarean section. In all pregnancies, neutrophilic infiltration was present at the maternal-fetal interface (decidua, placenta, fetal membranes), in various fetal tissues, and in fetal retina, choroid, and optic nerve (first trimester infection only). Consistent vertical transmission in this primate model may provide a platform to assess risk factors and test therapeutic interventions for interruption of fetal infection. The results may also suggest that maternal-fetal ZIKV transmission in human pregnancy may be more frequent than currently appreciated.


PLOS Neglected Tropical Diseases | 2016

Heterologous Protection against Asian Zika Virus Challenge in Rhesus Macaques

Matthew T. Aliota; Dawn M. Dudley; Christina M. Newman; Emma L. Mohr; Dane D. Gellerup; Meghan E. Breitbach; Connor R. Buechler; Mustafa N. Rasheed; Mariel S. Mohns; Andrea M. Weiler; Gabrielle L. Barry; Kim L. Weisgrau; Josh Eudailey; Eva G. Rakasz; Logan J. Vosler; Jennifer Post; Saverio Capuano; Thaddeus G. Golos; Sallie R. Permar; Jorge E. Osorio; Thomas C. Friedrich; Shelby L. O’Connor; David H. O’Connor

Background Zika virus (ZIKV; Flaviviridae, Flavivirus) was declared a public health emergency of international concern by the World Health Organization (WHO) in February 2016, because of the evidence linking infection with ZIKV to neurological complications, such as Guillain-Barre Syndrome in adults and congenital birth defects including microcephaly in the developing fetus. Because development of a ZIKV vaccine is a top research priority and because the genetic and antigenic variability of many RNA viruses limits the effectiveness of vaccines, assessing whether immunity elicited against one ZIKV strain is sufficient to confer broad protection against all ZIKV strains is critical. Recently, in vitro studies demonstrated that ZIKV likely circulates as a single serotype. Here, we demonstrate that immunity elicited by African lineage ZIKV protects rhesus macaques against subsequent infection with Asian lineage ZIKV. Methodology/Principal Findings Using our recently developed rhesus macaque model of ZIKV infection, we report that the prototypical ZIKV strain MR766 productively infects macaques, and that immunity elicited by MR766 protects macaques against heterologous Asian ZIKV. Furthermore, using next generation deep sequencing, we found in vivo restoration of a putative N-linked glycosylation site upon replication in macaques that is absent in numerous MR766 strains that are widely being used by the research community. This reversion highlights the importance of carefully examining the sequence composition of all viral stocks as well as understanding how passage history may alter a virus from its original form. Conclusions/Significance An effective ZIKV vaccine is needed to prevent infection-associated fetal abnormalities. Macaques whose immune responses were primed by infection with East African ZIKV were completely protected from detectable viremia when subsequently rechallenged with heterologous Asian ZIKV. Therefore, these data suggest that immunogen selection is unlikely to adversely affect the breadth of vaccine protection, i.e., any Asian ZIKV immunogen that protects against homologous challenge will likely confer protection against all other Asian ZIKV strains.


Nature Communications | 2017

Oropharyngeal mucosal transmission of Zika virus in rhesus macaques

Christina M. Newman; Dawn M. Dudley; Matthew T. Aliota; Andrea M. Weiler; Gabrielle L. Barry; Mariel S. Mohns; Meghan E. Breitbach; Laurel M. Stewart; Connor R. Buechler; Michael E. Graham; Jennifer Post; Nancy Schultz-Darken; Eric Peterson; Wendy Newton; Emma L. Mohr; Saverio Capuano; David H. O’Connor; Thomas C. Friedrich

Zika virus is present in urine, saliva, tears, and breast milk, but the transmission risk associated with these body fluids is currently unknown. Here we evaluate the risk of Zika virus transmission through mucosal contact in rhesus macaques. Application of high-dose Zika virus directly to the tonsils of three rhesus macaques results in detectable plasma viremia in all animals by 2 days post-exposure; virus replication kinetics are similar to those observed in animals infected subcutaneously. Three additional macaques inoculated subcutaneously with Zika virus served as saliva donors to assess the transmission risk from contact with oral secretions from an infected individual. Seven naive animals repeatedly exposed to donor saliva via the conjunctivae, tonsils, or nostrils did not become infected. Our results suggest that there is a risk of Zika virus transmission via the mucosal route, but that the risk posed by oral secretions from individuals with a typical course of Zika virus infection is low.Zika virus (ZIKV) is present in body fluids, including saliva, but transmission risk through mucosal contact is not well known. Here, the authors show that oropharyngeal mucosal infection of macaques with a high ZIKV dose results in viremia, but that transmission risk from saliva of infected animals is low.


Nature Communications | 2017

Infection via mosquito bite alters Zika virus tissue tropism and replication kinetics in rhesus macaques

Dawn M. Dudley; Christina M. Newman; Joseph Lalli; Laurel M. Stewart; Michelle R. Koenig; Andrea M. Weiler; Matthew Semler; Gabrielle L. Barry; Katie R. Zarbock; Mariel S. Mohns; Meghan E. Breitbach; Nancy Schultz-Darken; Eric Peterson; Wendy Newton; Emma L. Mohr; Saverio Capuano; Jorge E. Osorio; Shelby L. O’Connor; David H. O’Connor; Thomas C. Friedrich; Matthew T. Aliota

Mouse and nonhuman primate models now serve as useful platforms to study Zika virus (ZIKV) pathogenesis, candidate therapies, and vaccines, but they rely on needle inoculation of virus: the effects of mosquito-borne infection on disease outcome have not been explored in these models. Here we show that infection via mosquito bite delays ZIKV replication to peak viral loads in rhesus macaques. Importantly, in mosquito-infected animals ZIKV tissue distribution was limited to hemolymphatic tissues, female reproductive tract tissues, kidney, and liver, potentially emulating key features of human ZIKV infections, most of which are characterized by mild or asymptomatic disease. Furthermore, deep sequencing analysis reveals that ZIKV populations in mosquito-infected monkeys show greater sequence heterogeneity and lower overall diversity than in needle-inoculated animals. This newly developed system will be valuable for studying ZIKV disease because it more closely mimics human infection by mosquito bite than needle-based inoculations.Vector saliva can affect infectivity and pathogenesis of vector-borne viruses, but this hasn’t been studied for Zika virus infection. Here, Dudley et al. show that mosquito-mediated Zika infection of macaques results in altered replication kinetics and greater sequence heterogeneity.


Genome Biology | 2014

Whole genome sequencing of SIV-infected macaques identifies candidate loci that may contribute to host control of virus replication

Adam J. Ericsen; Gabriel J. Starrett; Justin M. Greene; Michael Lauck; Muthuswamy Raveendran; David Rio Deiros; Mariel S. Mohns; Nicolas Vince; Brian T Cain; Ngoc H Pham; Jason T. Weinfurter; Adam L. Bailey; Melisa L. Budde; Roger W. Wiseman; Richard A. Gibbs; Donna M. Muzny; Thomas C. Friedrich; Jeffrey Rogers; David H. O'Connor

BackgroundA small percentage of human immunodeficiency virus (HIV)-infected people and simian immunodeficiency virus (SIV)-infected macaques control virus replication without antiretroviral treatment. The major determinant of this control is host expression of certain major histocompatibility complex alleles. However, this association is incompletely penetrant, suggesting that additional loci modify the major histocompatibility complexs protective effect. Here, to identify candidate control-modifying loci, we sequence the genomes of 12 SIV-infected Mauritian cynomolgus macaques that experienced divergent viral load set points despite sharing the protective M1 major histocompatibility complex haplotype.ResultsOur genome-wide analysis of haplotype-level variation identifies seven candidate control-modifying loci on chromosomes 2, 3, 7, 8, 9, 10, and 14. The highest variant density marks the candidate on chromosome 7, which is the only control-modifying locus to comprise genes with known immunological function. Upon closer inspection, we found an allele for one of these genes, granzyme B, to be enriched in M1(+) controllers. Given its established role as a cytotoxic effector molecule that participates in CD8-mediated killing of virus-infected cells, we test the role of variation within gzmb in modifying SIV control by prospectively challenging M1(+) granzyme B-defined macaques.ConclusionsOur study establishes a framework for using whole genome sequencing to identify haplotypes that may contribute to complex clinical phenotypes. Further investigation into the immunogenetics underlying spontaneous HIV control may contribute to the rational design of a vaccine that prevents acquired immune deficiency syndrome.


PLOS ONE | 2018

Ocular and uteroplacental pathology in a macaque pregnancy with congenital Zika virus infection.

Emma L. Mohr; Lindsey N. Block; Christina M. Newman; Laurel M. Stewart; Michelle R. Koenig; Matthew Semler; Meghan E. Breitbach; Leandro B. C. Teixeira; Xiankun Zeng; Andrea M. Weiler; Gabrielle L. Barry; Troy H. Thoong; Gregory J. Wiepz; Dawn M. Dudley; Heather A. Simmons; Andres Mejia; Terry K. Morgan; M. Shahriar Salamat; Sarah Kohn; Kathleen M. Antony; Matthew T. Aliota; Mariel S. Mohns; Jennifer M. Hayes; Nancy Schultz-Darken; Michele L. Schotzko; Eric Peterson; Saverio Capuano; Jorge E. Osorio; Shelby L. O’Connor; Thomas C. Friedrich

Congenital Zika virus (ZIKV) infection impacts fetal development and pregnancy outcomes. We infected a pregnant rhesus macaque with a Puerto Rican ZIKV isolate in the first trimester. The pregnancy was complicated by preterm premature rupture of membranes (PPROM), intraamniotic bacterial infection and fetal demise 49 days post infection (gestational day 95). Significant pathology at the maternal-fetal interface included acute chorioamnionitis, placental infarcts, and leukocytoclastic vasculitis of the myometrial radial arteries. ZIKV RNA was disseminated throughout fetal tissues and maternal immune system tissues at necropsy, as assessed by quantitative RT-PCR for viral RNA. Replicating ZIKV was identified in fetal tissues, maternal uterus, and maternal spleen by fluorescent in situ hybridization for viral replication intermediates. Fetal ocular pathology included a choroidal coloboma, suspected anterior segment dysgenesis, and a dysplastic retina. This is the first report of ocular pathology and prolonged viral replication in both maternal and fetal tissues following congenital ZIKV infection in a rhesus macaque. PPROM followed by fetal demise and severe pathology of the visual system have not been described in macaque congenital ZIKV infection previously. While this case of ZIKV infection during pregnancy was complicated by bacterial infection with PPROM, the role of ZIKV on this outcome cannot be precisely defined, and further nonhuman primate studies will determine if increased risk for PPROM or other adverse pregnancy outcomes are associated with congenital ZIKV infection.


PLOS Pathogens | 2016

Microbial Translocation and Inflammation Occur in Hyperacute Immunodeficiency Virus Infection and Compromise Host Control of Virus Replication

Adam J. Ericsen; Michael Lauck; Mariel S. Mohns; Sarah R. DiNapoli; James P. Mutschler; Justin M. Greene; Jason T. Weinfurter; Gabrielle Lehrer-Brey; Trent M. Prall; Samantha M. Gieger; Connor R. Buechler; Kristin Crosno; Eric J. Peterson; Matthew R. Reynolds; Roger W. Wiseman; Benjamin J. Burwitz; Jacob D. Estes; Jonah B. Sacha; Thomas C. Friedrich; Jason M. Brenchley; David H. O’Connor

Within the first three weeks of human immunodeficiency virus (HIV) infection, virus replication peaks in peripheral blood. Despite the critical, causal role of virus replication in determining transmissibility and kinetics of progression to acquired immune deficiency syndrome (AIDS), there is limited understanding of the conditions required to transform the small localized transmitted founder virus population into a large and heterogeneous systemic infection. Here we show that during the hyperacute “pre-peak” phase of simian immunodeficiency virus (SIV) infection in macaques, high levels of microbial DNA transiently translocate into peripheral blood. This, heretofore unappreciated, hyperacute-phase microbial translocation was accompanied by sustained reduction of lipopolysaccharide (LPS)-specific antibody titer, intestinal permeability, increased abundance of CD4+CCR5+ T cell targets of virus replication, and T cell activation. To test whether increasing gastrointestinal permeability to cause microbial translocation would amplify viremia, we treated two SIV-infected macaque ‘elite controllers’ with a short-course of dextran sulfate sodium (DSS)–stimulating a transient increase in microbial translocation and a prolonged recrudescent viremia. Altogether, our data implicates translocating microbes as amplifiers of immunodeficiency virus replication that effectively undermine the host’s capacity to contain infection.


Science Translational Medicine | 2015

Durable sequence stability and bone marrow tropism in a macaque model of human pegivirus infection.

Adam L. Bailey; Michael Lauck; Mariel S. Mohns; Eric J. Peterson; Kerry Beheler; Kevin Brunner; Kristin Crosno; Andres Mejia; James P. Mutschler; Matthew Gehrke; Justin M. Greene; Adam J. Ericsen; Andrea M. Weiler; Gabrielle Lehrer-Brey; Thomas C. Friedrich; Samuel D. Sibley; Esper G. Kallas; Saverio Capuano; Jeffrey Rogers; Tony L. Goldberg; Heather A. Simmons; David H. O’Connor

A monkey model of human pegivirus infection provides new insights into the biology of this virus. An animal model for the “Good Boy Virus” Human pegivirus (HPgV, formerly called GB virus C) can protect HIV-infected individuals from developing AIDS, a phenomenon that has earned it the nickname of the “Good Boy Virus.” How HPgV imparts this protective effect remains a mystery, in part because no animal model of HPgV infection exists. Bailey and colleagues discovered viruses related to HPgV in wild baboons and showed that these viruses can infect laboratory macaque monkeys, providing unprecedented insights into the transmission, replication, and anatomical preference of HPgV with a view towards understanding HPgV-mediated protection from AIDS. Human pegivirus (HPgV)—formerly known as GB virus C and hepatitis G virus—is a poorly characterized RNA virus that infects about one-sixth of the global human population and is transmitted frequently in the blood supply. We create an animal model of HPgV infection by infecting macaque monkeys with a new simian pegivirus (SPgV) discovered in wild baboons. Using this model, we provide a high-resolution, longitudinal picture of SPgV viremia where the dose, route, and timing of infection are known. We detail the highly variable acute phase of SPgV infection, showing that the viral load trajectory early in infection is dependent on the infecting dose, whereas the chronic-phase viremic set point is not. We also show that SPgV has an extremely low propensity for accumulating sequence variation, with no consensus-level variants detected during the acute phase of infection and an average of only 1.5 variants generated per 100 infection-days. Finally, we show that SPgV RNA is highly concentrated in only two tissues: spleen and bone marrow, with bone marrow likely producing most of the virus detected in plasma. Together, these results reconcile several paradoxical observations from cross-sectional analyses of HPgV in humans and provide an animal model for studying pegivirus biology.


Journal of Virology | 2015

Expansion of Simian Immunodeficiency Virus (SIV)-Specific CD8 T Cell Lines from SIV-Naive Mauritian Cynomolgus Macaques for Adoptive Transfer

Mariel S. Mohns; Justin M. Greene; Brian T Cain; Ngoc H Pham; Emma Gostick; David A. Price; David H. O'Connor

ABSTRACT CD8 T cells play a crucial role in the control of human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV). However, the specific qualities and characteristics of an effective CD8 T cell response remain unclear. Although targeting breadth, cross-reactivity, polyfunctionality, avidity, and specificity are correlated with HIV control, further investigation is needed to determine the precise contributions of these various attributes to CD8 T cell efficacy. We developed protocols for isolating and expanding SIV-specific CD8 T cells from SIV-naive Mauritian cynomolgus macaques (MCM). These cells exhibited an effector memory phenotype, produced cytokines in response to cognate antigen, and suppressed viral replication in vitro. We further cultured cell lines specific for four SIV-derived epitopes, Nef103–111 RM9, Gag389–394 GW9, Env338–346 RF9, and Nef254–262 LT9. These cell lines were up to 94.4% pure, as determined by major histocompatibility complex (MHC) tetramer analysis. After autologous transfer into two MCM recipients, expanded CD8 T cells persisted in peripheral blood and lung tissue for at least 24 weeks and trafficked to multiple extralymphoid tissues. However, these cells did not impact the acute-phase SIV load after challenge compared to historic controls. The expansion and autologous transfer of SIV-specific T cells into naive animals provide a unique model for exploring cellular immunity and the control of SIV infection and facilitate a systematic evaluation of therapeutic adoptive transfer strategies for eradication of the latent reservoir. IMPORTANCE CD8 T cells play a crucial role in the control of human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV). Autologous adoptive transfer studies followed by SIV challenge may help define the critical elements of an effective T cell response to HIV and SIV infection. We developed protocols for isolating and expanding SIV-specific CD8 T cells from SIV-naive Mauritian cynomolgus macaques. This is an important first step toward the development of autologous transfer strategies to explore cellular immunity and potential therapeutic applications in the SIV model.

Collaboration


Dive into the Mariel S. Mohns's collaboration.

Top Co-Authors

Avatar

Thomas C. Friedrich

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Meghan E. Breitbach

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Christina M. Newman

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Dawn M. Dudley

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Emma L. Mohr

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Saverio Capuano

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Andrea M. Weiler

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Gabrielle L. Barry

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Matthew T. Aliota

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Nancy Schultz-Darken

University of Wisconsin-Madison

View shared research outputs
Researchain Logo
Decentralizing Knowledge