Network


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

Hotspot


Dive into the research topics where Evandro R. Winkelmann is active.

Publication


Featured researches published by Evandro R. Winkelmann.


Journal of Virology | 2009

Construction and Characterization of a Single-Cycle Chimeric Flavivirus Vaccine Candidate That Protects Mice against Lethal Challenge with Dengue Virus Type 2

Ryosuke Suzuki; Evandro R. Winkelmann; Peter W. Mason

ABSTRACT We have previously described a novel flavivirus vaccine technology based on a single-cycle, capsid (C) gene-deleted flavivirus called RepliVAX. RepliVAX can be propagated in cells that express high levels of C but undergoes only a single cycle of infection in vaccinated hosts. Here we report that we have adapted our RepliVAX technology to produce a dengue vaccine by replacing the prM/E genes of RepliVAX WN (a West Nile virus [WNV] RepliVAX) with the same genes of dengue virus type 2 (DENV2). Our first RepliVAX construct for dengue virus (RepliVAX D2) replicated poorly in WNV C-expressing cells. However, addition of mutations in prM and E that were selected during blind passage of a RepliVAX D2 derivative was used to produce a second-generation RepliVAX D2 (designated D2.2) that displayed acceptable growth in WNV C-expressing cells. RepliVAX D2.2 grew better in DENV2 C-expressing cells than WNV C-expressing cells, but after several passages in DENV2 C-expressing cells it acquired further mutations that permitted efficient growth in WNV C-expressing cells. We tested the potency and efficacy of RepliVAX D2.2 in a well-described immunodeficient mouse model for dengue (strain AG129; lacking the receptors for both type I and type II interferons). These mice produced dose-dependent DENV2-neutralizing antibody responses when vaccinated with RepliVAX D2.2. When challenged with 240 50% lethal doses of DENV2, mice given a single inoculation of RepliVAX D2.2 survived significantly longer than sham-vaccinated animals, although some of these severely immunocompromised mice eventually died from the challenge. Taken together these studies indicate that the RepliVAX technology shows promise for use in the development of vaccines that can be used to prevent dengue.


Vaccine | 2009

RepliVAX WN, a single-cycle flavivirus vaccine to prevent West Nile disease, elicits durable protective immunity in hamsters.

Douglas G. Widman; Tomohiro Ishikawa; Evandro R. Winkelmann; Ernesto Infante; Nigel Bourne; Peter W. Mason

West Nile virus (WNV) causes significant disease, yet no vaccines exist to prevent WN disease in humans. We have previously reported that RepliVAX WN is a safe and efficacious vaccine in mouse and hamster models of WN disease. Here, we report that vaccination of hamsters with RepliVAX WN induces antibody responses that remain stable for at least 6 months. Furthermore, animals challenged with virulent WNV 6 months after vaccination were protected from disease as well as those challenged 2 months post-vaccination, with no vaccinated animals succumbing to WNV challenge. These results indicate that RepliVAX is capable of inducing durable protective immunity after a single dose.


Vaccine | 2010

Immunogenicity of RepliVAX WN, a novel single-cycle West Nile virus vaccine.

Michelle H. Nelson; Evandro R. Winkelmann; Yinghong Ma; Jingya Xia; Peter W. Mason; Nigel Bourne; Gregg N. Milligan

We recently reported that immunization with RepliVAX WN, a single-cycle West Nile virus (WNV) vaccine, protected mice against WNV challenge. We have extended these studies by characterizing the RepliVAX WN-elicited antibody and T cell responses. WNV-specific IgG antibody responses comprised predominantly of IgG(2c) and IgG(2b) subclasses were detected 8 months after immunization. Vigorous WNV-specific CD4(+) and CD8(+) T cell responses directed at both structural and nonstructural WNV proteins were detected which were characterized by cytolytic activity and secretion of IFN-γ and TNF-α. Importantly, RepliVAX WN immunization resulted in vigorous CD8(+) memory T cell responses detected at 8 months after immunization.


F1000Research | 2016

West Nile Virus Infection in the Central Nervous System

Evandro R. Winkelmann; Huanle Luo; Tian Wang

West Nile virus (WNV), a neurotropic single-stranded flavivirus has been the leading cause of arboviral encephalitis worldwide. Up to 50% of WNV convalescent patients in the United States were reported to have long-term neurological sequelae. Neither antiviral drugs nor vaccines are available for humans. Animal models have been used to investigate WNV pathogenesis and host immune response in humans. In this review, we will discuss recent findings from studies in animal models of WNV infection, and provide new insights on WNV pathogenesis and WNV-induced host immunity in the central nervous system.


Journal of Virology | 2013

TLR3- and MyD88-Dependent Signaling Differentially Influences the Development of West Nile Virus-Specific B Cell Responses in Mice following Immunization with RepliVAX WN, a Single-Cycle Flavivirus Vaccine Candidate

Jingya Xia; Evandro R. Winkelmann; Summer R. Gorder; Peter W. Mason; Gregg N. Milligan

ABSTRACT Recognition of conserved pathogen-associated molecular patterns (PAMPs) by host pattern recognition receptors (PRRs) results in the activation of innate signaling pathways that drive the innate immune response and ultimately shape the adaptive immune response. RepliVAX WN, a single-cycle flavivirus (SCFV) vaccine candidate derived from West Nile virus (WNV), is intrinsically adjuvanted with multiple PAMPs and induces a vigorous anti-WNV humoral response. However, the innate mechanisms that link pattern recognition and development of vigorous antigen-specific B cell responses are not completely understood. Moreover, the roles of individual PRR signaling pathways in shaping the B cell response to this live attenuated SCFV vaccine have not been established. We examined and compared the role of TLR3- and MyD88-dependent signaling in the development of anti-WNV-specific antibody-secreting cell responses and memory B cell responses induced by RepliVAX WN. We found that MyD88 deficiency significantly diminished B cell responses by impairing B cell activation, development of germinal centers (GC), and the generation of long-lived plasma cells (LLPCs) and memory B cells (MBCs). In contrast, TLR3 deficiency had more effect on maintenance of GCs and development of LLPCs, whereas differentiation of MBCs was unaffected. Our data suggest that both TLR3- and MyD88-dependent signaling are involved in the intrinsic adjuvanting of RepliVAX WN and differentially contribute to the development of vigorous WNV-specific antibody and B cell memory responses following immunization with this novel SCFV vaccine.


Vaccine | 2011

Enhancing the utility of a prM/E-expressing chimeric vaccine for Japanese encephalitis by addition of the JEV NS1 gene

Tomohiro Ishikawa; Gongbo Wang; Douglas G. Widman; Ernesto Infante; Evandro R. Winkelmann; Nigel Bourne; Peter W. Mason

Recently, we demonstrated that a single-cycle West Nile virus (WNV) named RepliVAX WN could be used to produce a chimeric Japanese encephalitis (JE) vaccine (RepliVAX JE) by replacing the WNV prM/E genes with those of JEV. Here, we tested if replacement of WNV NS1 gene in RepliVAX JE with that of JEV (producing TripliVAX JE) could produce a superior vaccine. TripliVAX JE elicited higher anti-E immunity and displayed better efficacy in mice than RepliVAX JE. Furthermore, TripliVAX JE displayed reduced immune interference caused by pre-existing anti-NS1 immunity. Thus, we propose prM/E/NS1 chimerization as a new strategy for flavivirus vaccine development.


Journal of Virology | 2016

Dysregulation of Toll-Like Receptor 7 Compromises Innate and Adaptive T Cell Responses and Host Resistance to an Attenuated West Nile Virus Infection in Old Mice

Guorui Xie; Huanle Luo; Lan Pang; Bi Hung Peng; Evandro R. Winkelmann; Brenna McGruder; Joseph Hesse; Melissa C. Whiteman; Gerald A. Campbell; Gregg N. Milligan; Yingzi Cong; Alan D. T. Barrett; Tian Wang

ABSTRACT The elderly are known to have enhanced susceptibility to infections and an impaired capacity to respond to vaccination. West Nile virus (WNV), a mosquito-borne flavivirus, has induced severe neurological symptoms, mostly in the elderly population. No vaccines are available for human use. Recent work showed that an attenuated WNV, a nonstructural (NS) 4B-P38G mutant, induced no lethality but strong immune responses in young (6- to 10-week-old) mice. While studying protective efficacy, we found unexpectedly that old (21- to 22-month) mice were susceptible to WNV NS4B-P38G mutant infection but were protected from subsequent lethal wild-type WNV challenge. Compared to responses in young mice, the NS4B-P38G mutant triggered higher inflammatory cytokine and interleukin-10 (IL-10) production, a delayed γδ T cell expansion, and lower antibody and WNV-specific T cell responses in old mice. Toll-like receptor 7 (TLR7) is expressed on multiple types of cells. Impaired TLR7 signaling in old mice led to dendritic cell (DC) antigen-presenting function compromise and a reduced γδ T cell and regulatory T cell (Treg) expansion during NS4B-P38G mutant infection. R848, a TLR7 agonist, decreased host vulnerability in NS4B-P38G-infected old mice by enhancing γδ T cell and Treg expansion and the antigen-presenting capacity of DCs, thereby promoting T cell responses. In summary, our results suggest that dysregulation of TLR7 partially contributes to impaired innate and adaptive T cell responses and an enhanced vulnerability in old mice during WNV NS4B-P38G mutant infection. R848 increases the safety and efficacy during immunization of old mice with the WNV NS4B-P38G mutant. IMPORTANCE The elderly are known to have enhanced susceptibility to infections and an impaired capacity to respond to vaccination. West Nile virus (WNV), an emerging mosquito-borne flavivirus, has induced severe neurological symptoms more frequently in the elderly population. No vaccines are available for human use. Here, we used an aged mouse model to investigate the protective efficacy of an attenuated WNV, the nonstructural 4B-P38G mutant, which was previously shown to induce no lethality but strong immune responses in young adult mice. Studies that contribute to a mechanistic understanding of immune defects in the elderly will allow the development of strategies to improve responses to infectious diseases and to increase vaccine efficacy and safety in aging individuals.


Vaccine | 2012

Intrinsic adjuvanting of a novel single-cycle flavivirus vaccine in the absence of type I interferon receptor signaling

Evandro R. Winkelmann; Douglas G. Widman; Jingya Xia; Tomohiro Ishikawa; Mindy Miller-Kittrell; Michelle H. Nelson; Nigel Bourne; Frank Scholle; Peter W. Mason; Gregg N. Milligan

Type I interferons (IFNs) are critical for controlling pathogenic virus infections and can enhance immune responses. Hence their impact on the effectiveness of live-attenuated vaccines involves a balance between limiting viral antigen expression and enhancing the development of adaptive immune responses. We examined the influence of type I IFNs on these parameters following immunization with RepliVAX WN, a single-cycle flavivirus vaccine (SCFV) against West Nile virus (WNV) disease. RepliVAX WN-immunized mice produced IFN-α and displayed increased IFN-stimulated gene transcription in draining lymph nodes (LN). SCFV gene expression was over 100 fold-higher on days 1-3 post-infection in type I IFN receptor knockout mice (IFNAR(-/-)) compared to wild-type (wt) mice indicating a profound IFN-mediated suppression of SCFV gene expression in the wt animals. IFNAR(-/-) mice produced nearly equivalent levels of WNV-specific serum IgG and WNV-specific CD4(+) T cell responses compared to wt mice. However, significantly higher numbers of WNV-specific CD8(+) T cells were produced by IFNAR(-/-) mice and a significantly greater percentage of these T cells from IFNAR(-/-) mice produced only IFN-γ following antigen-specific re-stimulation. This altered cytokine expression was not associated with increased antigen load suggesting the loss of type I IFN receptor signaling was responsible for the altered quality of the CD8(+) effector T cell response. Together, these results indicate that although type I IFN is not essential for the intrinsic adjuvanting of RepliVAX WN, it plays a role in shaping the cytokine secretion profiles of CD8(+) effector T cells elicited by this SCFV.


Virology | 2014

Subcapsular sinus macrophages limit dissemination of West Nile virus particles after inoculation but are not essential for the development of West Nile virus-specific T cell responses

Evandro R. Winkelmann; Douglas G. Widman; Jingya Xia; Alison J. Johnson; Nico van Rooijen; Peter W. Mason; Nigel Bourne; Gregg N. Milligan

Macrophages encounter flaviviruses early after injection by arthropod vectors. Using in vivo imaging of mice inoculated with firefly luciferase-expressing single-cycle flavivirus particles (FLUC-SCFV), we examined the initial dissemination of virus particles in the presence or absence of lymph node (LN)-resident macrophages. Higher luciferase activity, indicating higher SCFV gene expression, was detected in the footpad of macrophage-depleted mice after 24h post infection (hpi). Moreover, FLUC-SCFV particles disseminated to the spleen within 14 hpi in macrophage-depleted, but not control mice. Although macrophages presented SCFV to naïve T cells in vitro, depletion of subcapsular sinus (SCS) macrophages did not alter the magnitude or effector function of the WNV-specific CD8(+) T cell response. Together, these results indicate that SCS macrophages play a role in limiting the dissemination of SCFV early in infection but are not required for the generation of a polyfunctional WNV-specific CD8(+) T cell response in the draining LN.


Antiviral Research | 2018

Zika, dengue and yellow fever viruses induce differential anti-viral immune responses in human monocytic and first trimester trophoblast cells

Huanle Luo; Evandro R. Winkelmann; Ildefonso Fernández-Salas; Li Li; Sandra V. Mayer; Rogelio Danis-Lozano; Rosa Ma Sanchez-Casas; Nikos Vasilakis; Robert B. Tesh; Alan D. T. Barrett; Scott C. Weaver; Tian Wang

ABSTRACT Zika virus (ZIKV) is a mosquito‐borne flavivirus associated with severe neonatal birth defects, but the causative mechanism is incompletely understood. ZIKV shares sequence homology and early clinical manifestations with yellow fever virus (YFV) and dengue virus (DENV) and are all transmitted in urban cycles by the same species of mosquitoes. However, YFV and DENV have been rarely reported to cause congenital diseases. Here, we compared infection with a contemporary ZIKV strain (FSS13025) to YFV17D and DENV‐4 in human monocytic cells (THP‐1) and first‐trimester trophoblasts (HTR‐8). Our results suggest that all three viruses have similar tropisms for both cells. Nevertheless, ZIKV induced strong type 1 IFN and inflammatory cytokine and chemokine production in monocytes and peripheral blood mononuclear cells. Furthermore, ZIKV infection in trophoblasts induced lower IFN and higher inflammatory immune responses. Placental inflammation is known to contribute to the risk of brain damage in preterm newborns. Inhibition of toll‐like receptor (TLR)3 and TLR8 each abrogated the inflammatory cytokine responses in ZIKV‐infected trophoblasts. Our findings identify a potential link between maternal immune activation and ZIKV‐induced congenital diseases, and a potential therapeutic strategy that targets TLR‐mediated inflammatory responses in the placenta. HighlightsZIKV, YFV and DENV have similar tropisms for human monocytic cells and first‐trimester trophoblasts.ZIKV induced strong anti‐viral immune responses in human monocytes and peripheral blood mononuclear cells.ZIKV infection in trophoblasts induced lower IFN and higher inflammatory immune Responses.TLRs 3 and 8 mediate inflammatory cytokine responses in trophoblasts during ZIKV infection.

Collaboration


Dive into the Evandro R. Winkelmann's collaboration.

Top Co-Authors

Avatar

Gregg N. Milligan

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Huanle Luo

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Nigel Bourne

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Tian Wang

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Douglas G. Widman

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Jingya Xia

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Alan D. T. Barrett

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Guorui Xie

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Michelle H. Nelson

Medical University of South Carolina

View shared research outputs
Researchain Logo
Decentralizing Knowledge