Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Elife ; 72018 06 19.
Article in English | MEDLINE | ID: mdl-29914621

ABSTRACT

Type I interferon (IFN-I) responses are critical for the control of RNA virus infections, however, many viruses, including Dengue (DENV) and Chikungunya (CHIKV) virus, do not directly activate plasmacytoid dendritic cells (pDCs), robust IFN-I producing cells. Herein, we demonstrated that DENV and CHIKV infected cells are sensed by pDCs, indirectly, resulting in selective IRF7 activation and IFN-I production, in the absence of other inflammatory cytokine responses. To elucidate pDC immunomodulatory functions, we developed a mouse model in which IRF7 signaling is restricted to pDC. Despite undetectable levels of IFN-I protein, pDC-restricted IRF7 signaling controlled both viruses and was sufficient to protect mice from lethal CHIKV infection. Early pDC IRF7-signaling resulted in amplification of downstream antiviral responses, including an accelerated natural killer (NK) cell-mediated type II IFN response. These studies revealed the dominant, yet indirect role of pDC IRF7-signaling in directing both type I and II IFN responses during arbovirus infections.


Subject(s)
Chikungunya Fever/immunology , Dengue/immunology , Host-Pathogen Interactions/immunology , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-7/immunology , Interferon Type I/immunology , Animals , Chikungunya Fever/genetics , Chikungunya Fever/mortality , Chikungunya Fever/pathology , Chikungunya virus/growth & development , Chikungunya virus/immunology , Chikungunya virus/pathogenicity , Dendritic Cells/immunology , Dendritic Cells/virology , Dengue/genetics , Dengue/mortality , Dengue/pathology , Dengue Virus/growth & development , Dengue Virus/immunology , Dengue Virus/pathogenicity , Disease Models, Animal , Female , Gene Expression Regulation , Humans , Interferon Regulatory Factor-3/deficiency , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-7/deficiency , Interferon Regulatory Factor-7/genetics , Interferon Type I/genetics , Interferon-gamma/genetics , Interferon-gamma/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/virology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Viral/antagonists & inhibitors , RNA, Viral/genetics , RNA, Viral/immunology , Signal Transduction , Spleen/immunology , Spleen/virology , Survival Analysis
2.
J Virol ; 90(21): 9683-9692, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27535047

ABSTRACT

RNA viruses present an extraordinary threat to human health, given their sudden and unpredictable appearance, the potential for rapid spread among the human population, and their ability to evolve resistance to antiviral therapies. The recent emergence of chikungunya virus, Zika virus, and Ebola virus highlights the struggles to contain outbreaks. A significant hurdle is the availability of antivirals to treat the infected or protect at-risk populations. While several compounds show promise in vitro and in vivo, these outbreaks underscore the need to accelerate drug discovery. The replication of several viruses has been described to rely on host polyamines, small and abundant positively charged molecules found in the cell. Here, we describe the antiviral effects of two molecules that alter polyamine levels: difluoromethylornithine (DFMO; also called eflornithine), which is a suicide inhibitor of ornithine decarboxylase 1 (ODC1), and diethylnorspermine (DENSpm), an activator of spermidine/spermine N1-acetyltransferase (SAT1). We show that reducing polyamine levels has a negative effect on diverse RNA viruses, including several viruses involved in recent outbreaks, in vitro and in vivo These findings highlight the importance of the polyamine biosynthetic pathway to viral replication, as well as its potential as a target in the development of further antivirals or currently available molecules, such as DFMO. IMPORTANCE: RNA viruses present a significant hazard to human health, and combatting these viruses requires the exploration of new avenues for targeting viral replication. Polyamines, small positively charged molecules within the cell, have been demonstrated to facilitate infection for a few different viruses. Our study demonstrates that diverse RNA viruses rely on the polyamine pathway for replication and highlights polyamine biosynthesis as a promising drug target.


Subject(s)
Antiviral Agents/pharmacology , Polyamines/metabolism , RNA Viruses/drug effects , Acetyltransferases/metabolism , Animals , Cell Line , Chikungunya Fever/drug therapy , Chikungunya Fever/virology , Chikungunya virus/drug effects , Chikungunya virus/metabolism , Disease Outbreaks , Ebolavirus/drug effects , Ebolavirus/metabolism , Eflornithine/pharmacology , Hemorrhagic Fever, Ebola/drug therapy , Hemorrhagic Fever, Ebola/virology , Humans , Mice , Mice, Inbred C57BL , Spermine/analogs & derivatives , Spermine/pharmacology , Virus Replication/drug effects , Zika Virus/drug effects , Zika Virus Infection/drug therapy , Zika Virus Infection/virology
3.
J Exp Med ; 209(5): 1029-47, 2012 May 07.
Article in English | MEDLINE | ID: mdl-22508836

ABSTRACT

Autophagy is an important survival pathway and can participate in the host response to infection. Studying Chikungunya virus (CHIKV), the causative agent of a major epidemic in India, Southeast Asia, and southern Europe, we reveal a novel mechanism by which autophagy limits cell death and mortality after infection. We use biochemical studies and single cell multispectral assays to demonstrate that direct infection triggers both apoptosis and autophagy. CHIKV-induced autophagy is mediated by the independent induction of endoplasmic reticulum and oxidative stress pathways. These cellular responses delay apoptotic cell death by inducing the IRE1α-XBP-1 pathway in conjunction with ROS-mediated mTOR inhibition. Silencing of autophagy genes resulted in enhanced intrinsic and extrinsic apoptosis, favoring viral propagation in cultured cells. Providing in vivo evidence for the relevance of our findings, Atg16L(HM) mice, which display reduced levels of autophagy, exhibited increased lethality and showed a higher sensitivity to CHIKV-induced apoptosis. Based on kinetic studies and the observation that features of apoptosis and autophagy were mutually exclusive, we conclude that autophagy inhibits caspase-dependent cell death but is ultimately overwhelmed by viral replication. Our study suggests that inducers of autophagy may limit the pathogenesis of acute Chikungunya disease.


Subject(s)
Alphavirus Infections/physiopathology , Apoptosis/physiology , Autophagy/physiology , Endoplasmic Reticulum/physiology , Oxidative Stress/physiology , Signal Transduction/physiology , Animals , Autophagy-Related Proteins , Blotting, Western , Carrier Proteins/genetics , Caspases/metabolism , Cell Line , Chikungunya Fever , DNA-Binding Proteins/metabolism , Endoribonucleases/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Image Processing, Computer-Assisted , Mice , Mice, Mutant Strains , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Regulatory Factor X Transcription Factors , Transcription Factors/metabolism , X-Box Binding Protein 1
4.
PLoS Pathog ; 7(10): e1002322, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22028657

ABSTRACT

Chikungunya virus (CHIKV) is a re-emerging alphavirus that has caused significant disease in the Indian Ocean region since 2005. During this outbreak, in addition to fever, rash and arthritis, severe cases of CHIKV infection have been observed in infants. Challenging the notion that the innate immune response in infants is immature or defective, we demonstrate that both human infants and neonatal mice generate a robust type I interferon (IFN) response during CHIKV infection that contributes to, but is insufficient for, the complete control of infection. To characterize the mechanism by which type I IFNs control CHIKV infection, we evaluated the role of ISG15 and defined it as a central player in the host response, as neonatal mice lacking ISG15 were profoundly susceptible to CHIKV infection. Surprisingly, UbE1L⁻/⁻ mice, which lack the ISG15 E1 enzyme and therefore are unable to form ISG15 conjugates, displayed no increase in lethality following CHIKV infection, thus pointing to a non-classical role for ISG15. No differences in viral loads were observed between wild-type (WT) and ISG15⁻/⁻ mice, however, a dramatic increase in proinflammatory cytokines and chemokines was observed in ISG15⁻/⁻ mice, suggesting that the innate immune response to CHIKV contributes to their lethality. This study provides new insight into the control of CHIKV infection, and establishes a new model for how ISG15 functions as an immunomodulatory molecule in the blunting of potentially pathologic levels of innate effector molecules during the host response to viral infection.


Subject(s)
Alphavirus Infections/immunology , Chikungunya virus/physiology , Cytokines/immunology , Interferon Type I/immunology , Ubiquitin-Activating Enzymes/immunology , Ubiquitins/immunology , Alphavirus Infections/diagnosis , Alphavirus Infections/metabolism , Animals , Animals, Newborn , Chikungunya Fever , Chikungunya virus/pathogenicity , Cytokines/metabolism , Disease Models, Animal , Humans , Interferon Type I/metabolism , Mice , Mice, Inbred C57BL , Protein Binding , Recombination, Genetic , Retrospective Studies , Ubiquitin-Activating Enzymes/metabolism , Ubiquitins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...