Network


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

Hotspot


Dive into the research topics where Marion C. Lanteri is active.

Publication


Featured researches published by Marion C. Lanteri.


Journal of Clinical Investigation | 2009

Tregs control the development of symptomatic West Nile virus infection in humans and mice

Marion C. Lanteri; Katie M. O’Brien; Whitney E. Purtha; Mark J. Cameron; Jennifer M. Lund; Rachel E. Owen; John W. Heitman; Brian Custer; Dale F. Hirschkorn; Leslie H. Tobler; Nancy Kiely; Harry E. Prince; Lishomwa C. Ndhlovu; Douglas F. Nixon; Hany Kamel; David J. Kelvin; Michael P. Busch; Alexander Y. Rudensky; Michael S. Diamond; Philip J. Norris

West Nile virus (WNV) causes asymptomatic infection in most humans, but for undefined reasons, approximately 20% of immunocompetent individuals develop West Nile fever, a potentially debilitating febrile illness, and approximately 1% develop neuroinvasive disease syndromes. Notably, since its emergence in 1999, WNV has become the leading cause of epidemic viral encephalitis in North America. We hypothesized that CD4+ Tregs might be differentially regulated in subjects with symptomatic compared with those with asymptomatic WNV infection. Here, we show that in 32 blood donors with acute WNV infection, Tregs expanded significantly in the 3 months after index (RNA+) donations in all subjects. Symptomatic donors exhibited lower Treg frequencies from 2 weeks through 1 year after index donation yet did not show differences in systemic T cell or generalized inflammatory responses. In parallel prospective experimental studies, symptomatic WNV-infected mice also developed lower Treg frequencies compared with asymptomatic mice at 2 weeks after infection. Moreover, Treg-deficient mice developed lethal WNV infection at a higher rate than controls. Together, these results suggest that higher levels of peripheral Tregs after infection protect against severe WNV disease in immunocompetent animals and humans.


PLOS Pathogens | 2012

IL-1β signaling promotes CNS-intrinsic immune control of West Nile virus infection.

Hilario J. Ramos; Marion C. Lanteri; Gabriele Blahnik; Amina Negash; Mehul S. Suthar; Margaret M. Brassil; Khushbu Sodhi; Piper M. Treuting; Michael P. Busch; Philip J. Norris; Michael Gale

West Nile virus (WNV) is an emerging flavivirus capable of infecting the central nervous system (CNS) and mediating neuronal cell death and tissue destruction. The processes that promote inflammation and encephalitis within the CNS are important for control of WNV disease but, how inflammatory signaling pathways operate to control CNS infection is not defined. Here, we identify IL-1β signaling and the NLRP3 inflammasome as key host restriction factors involved in viral control and CNS disease associated with WNV infection. Individuals presenting with acute WNV infection displayed elevated levels of IL-1β in their plasma over the course of infection, suggesting a role for IL-1β in WNV immunity. Indeed, we found that in a mouse model of infection, WNV induced the acute production of IL-1β in vivo, and that animals lacking the IL-1 receptor or components involved in inflammasome signaling complex exhibited increased susceptibility to WNV pathogenesis. This outcome associated with increased accumulation of virus within the CNS but not peripheral tissues and was further associated with altered kinetics and magnitude of inflammation, reduced quality of the effector CD8+ T cell response and reduced anti-viral activity within the CNS. Importantly, we found that WNV infection triggers production of IL-1β from cortical neurons. Furthermore, we found that IL-1β signaling synergizes with type I IFN to suppress WNV replication in neurons, thus implicating antiviral activity of IL-1β within neurons and control of virus replication within the CNS. Our studies thus define the NLRP3 inflammasome pathway and IL-1β signaling as key features controlling WNV infection and immunity in the CNS, and reveal a novel role for IL-1β in antiviral action that restricts virus replication in neurons.


AIDS | 2010

HIV+ elite controllers have low HIV-specific T-cell activation yet maintain strong, polyfunctional T-cell responses.

Rachel E. Owen; John W. Heitman; Dale F. Hirschkorn; Marion C. Lanteri; Hope H. Biswas; Jeffrey N. Martin; Melissa R. Krone; Steven G. Deeks; Philip J. Norris

Objective:HIV+ elite controllers are a unique group of rare individuals who maintain undetectable viral loads in the absence of antiretroviral therapy. We studied immune responses in these individuals to inform vaccine development, with the goal of identifying the immune correlates of protection from HIV. Methods:We compared markers of cellular activation, HIV-specific immune responses and regulatory T (Treg) cell frequencies in four groups of individuals: HIV-negative healthy controls, elite controllers (HIV RNA level <75 copies/ml), individuals on HAART and individuals with HIV RNA level more than 10 000 copies/ml (noncontrollers). Results:Elite controllers possessed significantly lower levels of activated HIV-specific CD8+ T cells and of recently divided HIV-specific CD4+ T cells than noncontrollers, whereas these differences were not seen in the respective cytomegalovirus-specific T-cell populations. Elite controllers also mounted a stronger and broader cytokine and chemokine response following HIV-specific stimulation than individuals on HAART and noncontrollers. Finally, we found that HAART-suppressed individuals had elevated Treg cell frequencies, whereas elite controllers and noncontrollers maintained normal percentages of Treg cells. Conclusion:Elite controllers maintain high levels of HIV-specific immune responses with low levels of HIV-specific T-cell activation and do not have elevated Treg cell levels. Based on these data an ideal HIV vaccine would induce strong HIV-specific immune responses whereas minimizing HIV-specific T-cell activation.


The Lancet | 2017

An update on Zika virus infection

David Baud; Duane J. Gubler; Bruno Schaub; Marion C. Lanteri; Didier Musso

The epidemic history of Zika virus began in 2007, with its emergence in Yap Island in the western Pacific, followed in 2013-14 by a larger epidemic in French Polynesia, south Pacific, where the first severe complications and non-vector-borne transmission of the virus were reported. Zika virus emerged in Brazil in 2015 and was declared a national public health emergency after local researchers and physicians reported an increase in microcephaly cases. In 2016, WHO declared the recent cluster of microcephaly cases and other neurological disorders reported in Brazil a global public health emergency. Similar clusters of microcephaly cases were also observed retrospectively in French Polynesia in 2014. In 2015-16, Zika virus continued its spread to cause outbreaks in the Americas and the Pacific, and the first outbreaks were reported in continental USA, Africa, and southeast Asia. Non-vector-borne transmission was confirmed and Zika virus was established as a cause of severe neurological complications in fetuses, neonates, and adults. This Review focuses on important updates and gaps in the knowledge of Zika virus as of early 2017.


Cytometry Part A | 2009

The importance of Foxp3 antibody and fixation/permeabilization buffer combinations in identifying CD4+CD25+Foxp3+ regulatory T cells

Jacqueline P. Law; Dale F. Hirschkorn; Rachel E. Owen; Hope H. Biswas; Philip J. Norris; Marion C. Lanteri

Foxp3 is a key marker for CD4+ regulatory T cells (Tregs) and was used in developing a multiparameter flow cytometric panel to identify Tregs. Achieving reproducible staining and analysis first required optimization of Foxp3 staining. We present a comparative study of PCH101, 236A/E7, 3G3, 206D, 150D, and 259D/C7 clones of anti‐human‐Foxp3 antibodies used in combination with five different fixation/permeabilization buffers. Staining for CD25, CD152, and CD127 was also compared between fixation/permeabilization treatments. Promising antibody/buffer combinations were tested in a panel of peripheral blood mononuclear cells from 10 individuals, and then on fresh versus frozen cells from four individuals. Finally, different fluorochromes coupled to two representative antibodies were compared to optimize separation of Foxp3+ from Foxp3− events. Foxp3 gates were set using two gating strategies based on CD127+CD25− “non‐Tregs” or based on isotype controls. For Foxp3 staining, the best conditions for fixation/permeabilization were obtained using the eBioscience Foxp3, Imgenex, BioLegend, and BD Foxp3 buffers. Comparing results from 10 subjects, 259D/C7, PCH101, 236A/E7, and 206D antibodies yielded statistically higher levels of Foxp3 cells than those by 150D and 3G3 antibodies (mean = 6.9, 5.1, 4.7, and 3.7% compared with 1.7, and 0.3% of CD25+Foxp3+ events within CD4+ cells, respectively). Importantly, the “nonspecificity” of some antibodies observed with a Foxp3 gate based on isotype controls could be eliminated by setting the Foxp3 gate on “non‐Tregs”. Better separation of Foxp3+ and Foxp3− populations was observed using the PCH101 clone coupled to Alexa647 compared with FITC or the 259D/C7 clone coupled to PE compared with Alexa488 fluorochrome. Foxp3 staining can be highly variable and depends on the choice of antibody/buffer pair and the fluorochrome used. Selecting the correct population for setting the Foxp3 gate is critical to avoid including non‐Tregs in the Foxp3+ gate. The experiments presented here will aid in optimization of flow cytometry staining panels to quantify Treg frequencies in humans.


Transfusion | 2014

West Nile virus nucleic acid persistence in whole blood months after clearance in plasma: implication for transfusion and transplantation safety.

Marion C. Lanteri; Tzong-Hae Lee; Li Wen; Zhanna Kaidarova; Marjorie Bravo; Nancy Kiely; Hany Kamel; Leslie H. Tobler; Philip J. Norris; Michael P. Busch

Previous reports of West Nile virus (WNV) RNA persistence in blood compartments have raised concerns around the remaining risk of WNV transfusion transmission. This study characterized the dynamics of WNV viremia in blood compartments in a longitudinal cohort of 54 WNV‐infected blood donors.


PLOS ONE | 2017

Zika Virus Tissue and Blood Compartmentalization in Acute Infection of Rhesus Macaques.

Lark L. Coffey; Patricia A. Pesavento; Keesler Ri; Anil Singapuri; Watanabe J; Rie Watanabe; Yee J; Eliza Bliss-Moreau; Cruzen C; Christe Kl; Reader; von Morgenland W; Gibbons Am; Allen Am; Jeffrey M. Linnen; Kui Gao; Eric Delwart; Graham Simmons; Mars Stone; Marion C. Lanteri; Sonia Bakkour; M.P. Busch; John H. Morrison; Van Rompay Kk

Animal models of Zika virus (ZIKV) are needed to better understand tropism and pathogenesis and to test candidate vaccines and therapies to curtail the pandemic. Humans and rhesus macaques possess similar fetal development and placental biology that is not shared between humans and rodents. We inoculated 2 non-pregnant rhesus macaques with a 2015 Brazilian ZIKV strain. Consistent with most human infections, the animals experienced no clinical disease but developed short-lived plasma viremias that cleared as neutralizing antibody developed. In 1 animal, viral RNA (vRNA) could be detected longer in whole blood than in plasma. Despite no major histopathologic changes, many adult tissues contained vRNA 14 days post-infection with highest levels in hemolymphatic tissues. These observations warrant further studies to investigate ZIKV persistence and its potential clinical implications for transmission via blood products or tissue and organ transplants.


Transfusion | 2016

Zika virus: a new threat to the safety of the blood supply with worldwide impact and implications

Marion C. Lanteri; Steven H. Kleinman; Simone A. Glynn; Didier Musso; W. Keith Hoots; Brian Custer; Ester C. Sabino; Michael P. Busch

A n emerging pathogen is an infectious agent that was previously unknown or whose geographic range or virulence has expanded. Zika virus (ZIKV) qualifies as the latest emerging virus impacting the human population, with major implications for blood safety and availability. Here we review historical and recent outbreaks of ZIKV and consider the response of transfusion medicine experts to the current explosive ZIKV epidemic, in light of recent efforts to define processes and criteria for emerging infectious disease risk assessment and response measures related to blood safety.


The Journal of Infectious Diseases | 2008

Comprehensive Analysis of West Nile Virus–Specific T Cell Responses in Humans

Marion C. Lanteri; John W. Heitman; Rachel E. Owen; Thomas Busch; Nelly Gefter; Nancy Kiely; Hany Kamel; Leslie H. Tobler; Michael P. Busch; Philip J. Norris

BACKGROUND Cellular responses have been shown to play a role in immune control and clearance of West Nile virus (WNV) in murine models. However, little is known about the immunogenic regions of the virus or the phenotype of responding T cells in human infection. METHODS Frozen peripheral blood mononuclear cells (PBMCs) from 35 WNV-infected blood donors were screened for virus-specific T cell responses by an interferon-gamma (IFN-gamma) enzyme-linked immunosorbent spot assay that used 452 overlapping peptides spanning all WNV proteins. More-detailed phenotypic studies were performed on subjects with high-magnitude T cell responses. RESULTS In individuals with identified responses, the total number of recognized WNV peptides ranged from 1 to 9 (median, 2 peptides), and the overall magnitude of responses ranged from 50 to 4210 spot-forming cells (SFCs) per 10(6) PBMCs (median, 130 SFCs/10(6) PBMCs). A subset of 8 frequently recognized peptides from the regions of the genome encoding membrane, envelope, and nonstructural 3 and 4b proteins was identified. Phenotypic study of the highest magnitude WNV-specific T cell responses revealed that most were mediated by CD8+ cells that expressed perforin and/or granzyme B. CONCLUSIONS These findings are the first to define the breadth and characteristics of the human T cell response to WNV and have implications for candidate vaccine design and evaluation.


The Journal of Infectious Diseases | 2008

Interferon and Interferon-Induced Chemokine Expression Is Associated with Control of Acute Viremia in West Nile Virus-Infected Blood Donors

Leslie H. Tobler; Mark J. Cameron; Marion C. Lanteri; Harry E. Prince; Ali Danesh; Desmond Persad; Robert S. Lanciotti; Philip J. Norris; David J. Kelvin; Michael P. Busch

Abstract To understand early host responses controlling West Nile virus (WNV) infection, acutely viremic blood donors, identified by nucleic acid amplification testing, were enrolled and monitored for RNA-clearance and WNV-specific IgM and IgG antibodies. Viral load and chemokine and cytokine assays were performed on serial samples from donors whose index and first follow-up samples tested negative for IgM. A total of 84% of the specimens obtained from viremic donors before IgM/IgG seroconversion demonstrated a decreasing viral load. Levels of interferon (IFN)-α ere significantly increased before IgM seroconversion, relative to those in control specimens. CXCL10 and CCL2 were significantly elevated in donor specimens obtained before IgM seroconversion, compared with those obtained after IgM seroconversion. These findings suggest that IFN-mediated innate immunity plays a key role in initial control of WNV replication.

Collaboration


Dive into the Marion C. Lanteri's collaboration.

Top Co-Authors

Avatar

Michael P. Busch

Systems Research Institute

View shared research outputs
Top Co-Authors

Avatar

Philip J. Norris

Systems Research Institute

View shared research outputs
Top Co-Authors

Avatar

Didier Musso

Aix-Marseille University

View shared research outputs
Top Co-Authors

Avatar

Sheila M. Keating

Systems Research Institute

View shared research outputs
Top Co-Authors

Avatar

Lishomwa C. Ndhlovu

University of Hawaii at Manoa

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Brian Custer

Systems Research Institute

View shared research outputs
Top Co-Authors

Avatar

John W. Heitman

Systems Research Institute

View shared research outputs
Top Co-Authors

Avatar

Mars Stone

Systems Research Institute

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge