Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
2.
Nat Commun ; 14(1): 4481, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37491352

ABSTRACT

Inflammation in response to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection drives severity of coronavirus disease 2019 (COVID-19) and is influenced by host genetics. To understand mechanisms of inflammation, animal models that reflect genetic diversity and clinical outcomes observed in humans are needed. We report a mouse panel comprising the genetically diverse Collaborative Cross (CC) founder strains crossed to human ACE2 transgenic mice (K18-hACE2) that confers susceptibility to SARS-CoV-2. Infection of CC x K18-hACE2 resulted in a spectrum of survival, viral replication kinetics, and immune profiles. Importantly, in contrast to the K18-hACE2 model, early type I interferon (IFN-I) and regulated proinflammatory responses were required for control of SARS-CoV-2 replication in PWK x K18-hACE2 mice that were highly resistant to disease. Thus, virus dynamics and inflammation observed in COVID-19 can be modeled in diverse mouse strains that provide a genetically tractable platform for understanding anti-coronavirus immunity.


Subject(s)
COVID-19 , Interferon Type I , Humans , Mice , Animals , Cytokines , SARS-CoV-2 , Mice, Transgenic , Inflammation/genetics , Disease Models, Animal , Lung
3.
bioRxiv ; 2023 Apr 21.
Article in English | MEDLINE | ID: mdl-35233576

ABSTRACT

Inflammation in response to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection drives severity of coronavirus disease 2019 (COVID-19) and is influenced by host genetics. To understand mechanisms of inflammation, animal models that reflect genetic diversity and clinical outcomes observed in humans are needed. We report a mouse panel comprising the genetically diverse Collaborative Cross (CC) founder strains crossed to human ACE2 transgenic mice (K18-hACE2) that confers susceptibility to SARS-CoV-2. Infection of CC x K18- hACE2 resulted in a spectrum of survival, viral replication kinetics, and immune profiles. Importantly, in contrast to the K18-hACE2 model, early type I interferon (IFN-I) and regulated proinflammatory responses were required for control of SARS-CoV-2 replication in PWK x K18-hACE2 mice that were highly resistant to disease. Thus, virus dynamics and inflammation observed in COVID-19 can be modeled in diverse mouse strains that provide a genetically tractable platform for understanding anti-coronavirus immunity.

4.
J Immunol ; 209(2): 346-353, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35750336

ABSTRACT

Our recent data demonstrate a critical role of the RIG-I-like receptor family in regulating antifungal immunity against Aspergillus fumigatus in a murine model. However, the importance of this pathway in humans and the cell types that use this innate immune receptor family to detect A. fumigatus remain unresolved. In this study, using patients who underwent hematopoietic stem cell transplantation, we demonstrate that a polymorphism in human MAVS present in the donor genome was associated with the incidence of invasive pulmonary aspergillosis. Moreover, in a separate cohort of confirmed invasive pulmonary aspergillosis patients, polymorphisms in the IFIH1 gene alter the inflammatory response, including IFN-responsive chemokines. Returning to our murine model, we now demonstrate that CD11c+ Siglec F+ alveolar macrophages require Mavs expression to maintain host resistance against A. fumigatus. Our data support the role of MAVS signaling in mediating antifungal immunity in both mice and humans at least in part through the role of MAVS-dependent signaling in alveolar macrophages.


Subject(s)
Aspergillus fumigatus , Invasive Pulmonary Aspergillosis , Animals , Antifungal Agents , Disease Models, Animal , Humans , Macrophages, Alveolar , Mice
5.
Immunity ; 55(3): 382-384, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35263563

ABSTRACT

Macrophage activation is essential for effective immunity to infection but can also contribute to disease through incompletely understood mechanisms. In this issue of Immunity, Simpson et al. reveal that death of activated macrophages integrates extrinsic and intrinsic pathways of apoptosis that contribute to damaging host responses.


Subject(s)
Interferon-gamma , Macrophage Activation , Apoptosis , Caspase 8/metabolism , Cell Death , Interferon-gamma/metabolism , Ligands , Macrophages/immunology
6.
J Exp Med ; 219(2)2022 02 07.
Article in English | MEDLINE | ID: mdl-34889942

ABSTRACT

In addition to providing partial protection against pediatric tuberculosis, vaccination with bacille Calmette-Guérin (BCG) has been reported to confer nonspecific resistance to unrelated pulmonary pathogens, a phenomenon attributed to the induction of long-lasting alterations within the myeloid cell compartment. Here, we demonstrate that intravenous, but not subcutaneous, inoculation of BCG protects human-ACE2 transgenic mice against lethal challenge with SARS-CoV-2 (SCV2) and results in reduced viral loads in non-transgenic animals infected with an α variant. The observed increase in host resistance was associated with reductions in SCV2-induced tissue pathology, inflammatory cell recruitment, and cytokine production that multivariate analysis revealed as only partially related to diminished viral load. We propose that this protection stems from BCG-induced alterations in the composition and function of the pulmonary cellular compartment that impact the innate response to the virus and ensuing immunopathology. While intravenous BCG vaccination is not a clinically acceptable practice, our findings provide an experimental model for identifying mechanisms by which nonspecific stimulation of the pulmonary immune response promotes host resistance to SCV2 lethality.


Subject(s)
BCG Vaccine/immunology , COVID-19/immunology , SARS-CoV-2/immunology , Administration, Intravenous , Angiotensin-Converting Enzyme 2/metabolism , Animals , Chemokines/metabolism , Humans , Inflammation/pathology , Mice, Inbred C57BL , Mice, Transgenic , Viral Load
7.
PLoS Pathog ; 17(12): e1009678, 2021 12.
Article in English | MEDLINE | ID: mdl-34855915

ABSTRACT

Kyasanur Forest disease virus (KFDV) and the closely related Alkhurma hemorrhagic disease virus (AHFV) are emerging flaviviruses that cause severe viral hemorrhagic fevers in humans. Increasing geographical expansion and case numbers, particularly of KFDV in southwest India, class these viruses as a public health threat. Viral pathogenesis is not well understood and additional vaccines and antivirals are needed to effectively counter the impact of these viruses. However, current animal models of KFDV pathogenesis do not accurately reproduce viral tissue tropism or clinical outcomes observed in humans. Here, we show that pigtailed macaques (Macaca nemestrina) infected with KFDV or AHFV develop viremia that peaks 2 to 4 days following inoculation. Over the course of infection, animals developed lymphocytopenia, thrombocytopenia, and elevated liver enzymes. Infected animals exhibited hallmark signs of human disease characterized by a flushed appearance, piloerection, dehydration, loss of appetite, weakness, and hemorrhagic signs including epistaxis. Virus was commonly present in the gastrointestinal tract, consistent with human disease caused by KFDV and AHFV where gastrointestinal symptoms (hemorrhage, vomiting, diarrhea) are common. Importantly, RNAseq of whole blood revealed that KFDV downregulated gene expression of key clotting factors that was not observed during AHFV infection, consistent with increased severity of KFDV disease observed in this model. This work characterizes a nonhuman primate model for KFDV and AHFV that closely resembles human disease for further utilization in understanding host immunity and development of antiviral countermeasures.


Subject(s)
Disease Models, Animal , Encephalitis Viruses, Tick-Borne/pathogenicity , Encephalitis, Tick-Borne/virology , Hemorrhagic Fevers, Viral/virology , Macaca nemestrina , Animals , Chlorocebus aethiops , Cytokines/blood , Encephalitis Viruses, Tick-Borne/genetics , Encephalitis Viruses, Tick-Borne/immunology , Encephalitis, Tick-Borne/immunology , Encephalitis, Tick-Borne/pathology , Female , HEK293 Cells , Hemorrhagic Fevers, Viral/immunology , Hemorrhagic Fevers, Viral/pathology , Humans , Lymph Nodes/virology , Vero Cells , Viremia
8.
Cell Rep ; 37(4): 109888, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34706234

ABSTRACT

Dysregulated inflammation dominated by chemokine expression is a key feature of disease following infection with the globally important human pathogens Zika virus (ZIKV) and dengue virus, but a mechanistic understanding of how pro-inflammatory responses are initiated is lacking. Mitophagy is a quality-control mechanism that regulates innate immune signaling and cytokine production through selective degradation of damaged mitochondria. Here, we demonstrate that ZIKV nonstructural protein 5 (NS5) antagonizes mitophagy by binding to the host protein Ajuba and preventing its translocation to depolarized mitochondria where it is required for PINK1 activation and downstream signaling. Consequent mitophagy suppression amplifies the production of pro-inflammatory chemokines through protein kinase R (PKR) sensing of mitochondrial RNA. In Ajuba-/- mice, ZIKV induces early expression of pro-inflammatory chemokines associated with significantly enhanced dissemination to tissues. This work identifies Ajuba as a critical regulator of mitophagy and demonstrates a role for mitophagy in limiting systemic inflammation following infection by globally important human viruses.


Subject(s)
LIM Domain Proteins/metabolism , Mitophagy , Protein Kinases/metabolism , Signal Transduction , Zika Virus Infection/metabolism , Zika Virus/metabolism , eIF-2 Kinase/metabolism , A549 Cells , Animals , Chlorocebus aethiops , HEK293 Cells , HeLa Cells , Humans , LIM Domain Proteins/genetics , Mice , Mice, Knockout , Protein Kinases/genetics , Vero Cells , Zika Virus/genetics , Zika Virus Infection/genetics , eIF-2 Kinase/genetics
9.
bioRxiv ; 2021 Aug 31.
Article in English | MEDLINE | ID: mdl-34494021

ABSTRACT

Early events in the host response to SARS-CoV-2 are thought to play a major role in determining disease severity. During pulmonary infection, the virus encounters both myeloid and epithelioid lineage cells that can either support or restrict pathogen replication as well as respond with host protective versus detrimental mediators. In addition to providing partial protection against pediatric tuberculosis, vaccination with bacille Calmette-Guérin (BCG) has been reported to confer non-specific resistance to unrelated pulmonary pathogens, a phenomenon attributed to the induction of long-lasting alterations within the myeloid cell compartment. Here we demonstrate that prior intravenous, but not subcutaneous, administration of BCG protects human-ACE2 transgenic mice against lethal challenge with SARS-CoV-2 and results in reduced viral loads in non-transgenic animals infected with an alpha variant. The observed increase in host resistance was associated with reductions in SARS-CoV-2-induced tissue pathology, inflammatory cell recruitment and cytokine production that multivariate analysis revealed to be only partially related to diminished viral load. We propose that this protection stems from BCG-induced alterations in the composition and function of the pulmonary cellular compartment that impact the innate response to the virus and the ensuing immunopathology.

10.
Nature ; 585(7825): 414-419, 2020 09.
Article in English | MEDLINE | ID: mdl-32641828

ABSTRACT

Zika virus (ZIKV) belongs to the family Flaviviridae, and is related to other viruses that cause human diseases. Unlike other flaviviruses, ZIKV infection can cause congenital neurological disorders and replicates efficiently in reproductive tissues1-3. Here we show that the envelope protein (E) of ZIKV is polyubiquitinated by the E3 ubiquitin ligase TRIM7 through Lys63 (K63)-linked polyubiquitination. Accordingly, ZIKV replicates less efficiently in the brain and reproductive tissues of Trim7-/- mice. Ubiquitinated E is present on infectious virions of ZIKV when they are released from specific cell types, and enhances virus attachment and entry into cells. Specifically, K63-linked polyubiquitin chains directly interact with the TIM1 (also known as HAVCR1) receptor of host cells, which enhances virus entry in cells as well as in brain tissue in vivo. Recombinant ZIKV mutants that lack ubiquitination are attenuated in human cells and in wild-type mice, but not in live mosquitoes. Monoclonal antibodies against K63-linked polyubiquitin specifically neutralize ZIKV and reduce viraemia in mice. Our results demonstrate that the ubiquitination of ZIKV E is an important determinant of virus entry, tropism and pathogenesis.


Subject(s)
Ubiquitination , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Virus Internalization , Zika Virus/metabolism , Zika Virus/pathogenicity , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Brain/metabolism , Cell Line , Culicidae/cytology , Culicidae/virology , Endosomes/metabolism , Female , Hepatitis A Virus Cellular Receptor 1/metabolism , Humans , Male , Membrane Fusion , Mice , Organ Specificity , Polyubiquitin/immunology , Polyubiquitin/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Viral Tropism , Viremia/immunology , Viremia/prevention & control , Viremia/virology , Virus Replication , Zika Virus/chemistry , Zika Virus/genetics , Zika Virus Infection/prevention & control , Zika Virus Infection/virology
11.
mBio ; 10(6)2019 12 03.
Article in English | MEDLINE | ID: mdl-31796542

ABSTRACT

Flaviviruses are controlled by adaptive immune responses but are exquisitely sensitive to interferon-stimulated genes (ISGs). How coinfections, particularly simian immunodeficiency viruses (SIVs), that induce robust ISG signatures influence flavivirus clearance and pathogenesis is unclear. Here, we studied how Zika virus (ZIKV) infection is modulated in SIV-infected nonhuman primates. We measured ZIKV replication, cellular ZIKV RNA levels, and immune responses in non-SIV-infected and SIV-infected rhesus macaques (RMs), which we infected with ZIKV. Coinfected animals had a 1- to 2-day delay in peak ZIKV viremia, which was 30% of that in non-SIV-infected animals. However, ZIKV viremia was significantly prolonged in SIV-positive (SIV+) RMs. ISG levels at the time of ZIKV infection were predictive for lower ZIKV viremia in the SIV+ RMs, while prolonged ZIKV viremia was associated with muted and delayed adaptive responses in SIV+ RMs.IMPORTANCE Immunocompromised individuals often become symptomatic with infections which are normally fairly asymptomatic in healthy individuals. The particular mechanisms that underlie susceptibility to coinfections in human immunodeficiency virus (HIV)-infected individuals are multifaceted. ZIKV and other flaviviruses are sensitive to neutralizing antibodies, whose production can be limited in HIV-infected individuals but are also sensitive to type I interferons, which are expressed at high levels in HIV-infected individuals. Data in this study highlight how individual components of the innate and adaptive immune responses which become perturbed in HIV-infected individuals influence ZIKV infection.


Subject(s)
Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/immunology , Zika Virus Infection/immunology , Zika Virus Infection/virology , Zika Virus/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antiviral Agents/immunology , Female , Interferons/immunology , Macaca mulatta/immunology , Macaca mulatta/virology , Male , Viral Load/immunology , Viremia/immunology , Viremia/virology , Virus Replication/immunology
12.
Cell Rep ; 27(11): 3269-3283.e6, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31189110

ABSTRACT

Tripartite motif-containing protein 5α (TRIM5α) is a cellular antiviral restriction factor that prevents early events in retrovirus replication. The activity of TRIM5α is thought to be limited to retroviruses as a result of highly specific interactions with capsid lattices. In contrast to this current understanding, we show that both human and rhesus macaque TRIM5α suppress replication of specific flaviviruses. Multiple viruses in the tick-borne encephalitis complex are sensitive to TRIM5α-dependent restriction, but mosquito-borne flaviviruses, including yellow fever, dengue, and Zika viruses, are resistant. TRIM5α suppresses replication by binding to the viral protease NS2B/3 to promote its K48-linked ubiquitination and proteasomal degradation. Importantly, TRIM5α contributes to the antiviral function of IFN-I against sensitive flaviviruses in human cells. Thus, TRIM5α possesses remarkable plasticity in the recognition of diverse virus families, with the potential to influence human susceptibility to emerging flaviviruses of global concern.


Subject(s)
Flavivirus Infections/metabolism , Peptide Hydrolases/metabolism , Proteasome Endopeptidase Complex/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Viral Proteins/metabolism , Virus Replication , Animals , Antiviral Restriction Factors , Cats , Chlorocebus aethiops , Dendritic Cells/metabolism , Dendritic Cells/virology , Flavivirus/pathogenicity , Flavivirus/physiology , Flavivirus Infections/virology , HEK293 Cells , Humans , Protein Binding , Proteolysis , Substrate Specificity , Ubiquitination , Vero Cells
13.
PLoS Negl Trop Dis ; 12(7): e0006628, 2018 07.
Article in English | MEDLINE | ID: mdl-30011277

ABSTRACT

Crimean-Congo hemorrhagic fever (CCHF) is an acute, often fatal viral disease characterized by rapid onset of febrile symptoms followed by hemorrhagic manifestations. The etiologic agent, CCHF orthonairovirus (CCHFV), can infect several mammals in nature but only seems to cause clinical disease in humans. Over the past two decades there has been an increase in total number of CCHF case reports, including imported CCHF patients, and an expansion of CCHF endemic areas. Despite its increased public health burden there are currently no licensed vaccines or treatments to prevent CCHF. We here report the development and assessment of the protective efficacy of an adenovirus (Ad)-based vaccine expressing the nucleocapsid protein (N) of CCHFV (Ad-N) in a lethal immunocompromised mouse model of CCHF. The results show that Ad-N can protect mice from CCHF mortality and that this platform should be considered for future CCHFV vaccine strategies.


Subject(s)
Hemorrhagic Fever Virus, Crimean-Congo/immunology , Hemorrhagic Fever, Crimean/prevention & control , Nucleocapsid Proteins/immunology , Viral Vaccines/immunology , Animals , Antibodies, Viral/immunology , Disease Models, Animal , Female , Hemorrhagic Fever Virus, Crimean-Congo/genetics , Hemorrhagic Fever, Crimean/immunology , Hemorrhagic Fever, Crimean/virology , Humans , Male , Mice , Mice, Inbred C57BL , Nucleocapsid Proteins/administration & dosage , Nucleocapsid Proteins/genetics , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
14.
J Virol ; 92(17)2018 09 01.
Article in English | MEDLINE | ID: mdl-29925658

ABSTRACT

Chikungunya virus (CHIKV) is a mosquito-borne alphavirus that has evolved effective mechanisms to counteract the type I interferon (IFN) response. Upon recognition of the virus, cells secrete IFNs, which signal through transmembrane receptors (IFNAR) to phosphorylate STAT proteins (pSTAT). pSTAT dimers are transported into the nucleus by importin-α5 and activate the transcription of IFN-stimulated genes (ISGs), increasing cellular resistance to infection. Subsequently, STAT proteins are shuttled back into the cytoplasm by the exportin CRM1. CHIKV nonstructural protein 2 (nsP2) reduces ISG expression by inhibiting general host cell transcription and by specifically reducing the levels of nuclear pSTAT1 via an unknown mechanism. To systematically examine where nsP2 acts within the JAK/STAT signaling cascade, we used two well-characterized mutants of nsP2, P718S and KR649AA. Both mutations abrogate nsP2's ability to shut off host transcription, but only the KR649AA mutant localizes exclusively to the cytoplasm and no longer specifically inhibits JAK/STAT signaling. These mutant nsP2 proteins did not differentially affect IFNAR expression levels or STAT1 phosphorylation in response to IFNs. Coimmunoprecipitation experiments showed that in the presence of nsP2, STAT1 still effectively bound importin-α5. Chemically blocking CRM1-mediated nuclear export in the presence of nsP2 additionally showed that nuclear translocation of STAT1 is not affected by nsP2. nsP2 putatively has five domains. Redirecting the nsP2 KR649AA mutant or just nsP2's C-terminal methyltransferase-like domain into the nucleus strongly reduced nuclear pSTAT in response to IFN stimulation. This demonstrates that the C-terminal domain of nuclear nsP2 specifically inhibits the IFN response by promoting the nuclear export of STAT1.IMPORTANCE Chikungunya virus is an emerging pathogen associated with large outbreaks on the African, Asian, European, and both American continents. In most patients, infection results in high fever, rash, and incapacitating (chronic) arthralgia. CHIKV effectively inhibits the first line of defense, the innate immune response. As a result, stimulation of the innate immune response with interferons (IFNs) is ineffective as a treatment for CHIKV disease. The IFN response requires an intact downstream signaling cascade called the JAK/STAT signaling pathway, which is effectively inhibited by CHIKV nonstructural protein 2 (nsP2) via an unknown mechanism. The research described here specifies where in the JAK/STAT signaling cascade the IFN response is inhibited and which protein domain of nsP2 is responsible for IFN inhibition. The results illuminate new aspects of antiviral defense and CHIKV counterdefense strategies and will direct the search for novel antiviral compounds.


Subject(s)
Chikungunya virus/enzymology , Cysteine Endopeptidases/genetics , Immunity, Innate , Interferon Type I/antagonists & inhibitors , STAT1 Transcription Factor/genetics , Cell Line , Chikungunya virus/genetics , Chikungunya virus/physiology , Computer Simulation , Cysteine Endopeptidases/metabolism , HEK293 Cells , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immunoprecipitation , Interferon Type I/genetics , Interferon Type I/immunology , Methyltransferases/metabolism , Mutation , Signal Transduction/genetics , Signal Transduction/immunology , Virus Replication
15.
Cell Host Microbe ; 23(5): 672-685.e6, 2018 05 09.
Article in English | MEDLINE | ID: mdl-29746837

ABSTRACT

Progress toward understanding Zika virus (ZIKV) pathogenesis is hindered by lack of immunocompetent small animal models, in part because ZIKV fails to effectively antagonize Stat2-dependent interferon (IFN) responses in mice. To address this limitation, we first passaged an African ZIKV strain (ZIKV-Dak-41525) through Rag1-/- mice to obtain a mouse-adapted virus (ZIKV-Dak-MA) that was more virulent than ZIKV-Dak-41525 in mice treated with an anti-Ifnar1 antibody. A G18R substitution in NS4B was the genetic basis for the increased replication, and resulted in decreased IFN-ß production, diminished IFN-stimulated gene expression, and the greater brain infection observed with ZIKV-Dak-MA. To generate a fully immunocompetent mouse model of ZIKV infection, human STAT2 was introduced into the mouse Stat2 locus (hSTAT2 KI). Subcutaneous inoculation of pregnant hSTAT2 KI mice with ZIKV-Dak-MA resulted in spread to the placenta and fetal brain. An immunocompetent mouse model of ZIKV infection may prove valuable for evaluating countermeasures to limit disease.


Subject(s)
Mice/immunology , Zika Virus Infection/immunology , Zika Virus/immunology , Zika Virus/pathogenicity , Animals , Brain , Cell Survival , Disease Models, Animal , Female , Fetal Diseases/metabolism , Fetal Diseases/virology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immunity , Infectious Disease Transmission, Vertical , Interferon-beta/metabolism , Interferons/metabolism , Mice/genetics , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Placenta/metabolism , Pregnancy , Pregnancy Complications, Infectious/virology , RNA Helicases/genetics , Receptor, Interferon alpha-beta , STAT2 Transcription Factor/metabolism , Serine Endopeptidases/genetics , Viral Nonstructural Proteins/genetics , Zika Virus/genetics , Zika Virus Infection/virology
16.
J Immunol ; 200(2): 471-476, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29246952

ABSTRACT

Inflammatory monocyte (iMO) recruitment to the brain is a hallmark of many neurologic diseases. Prior to entering the brain, iMOs must egress into the blood from the bone marrow through a mechanism, which for known encephalitic viruses, is CCR2 dependent. In this article, we show that during La Crosse Virus-induced encephalitis, egress of iMOs was surprisingly independent of CCR2, with similar percentages of iMOs in the blood and brain of heterozygous and CCR2-/- mice following infection. Interestingly, CCR2 was required for iMO trafficking from perivascular areas to sites of virus infection within the brain. Thus, CCR2 was not essential for iMO trafficking to the blood or the brain but was essential for trafficking within the brain parenchyma. Analysis of other orthobunyaviruses showed that Jamestown Canyon virus also induced CCR2-independent iMO egress to the blood. These studies demonstrate that the CCR2 requirement for iMO egress to the blood is not universal for all viruses.


Subject(s)
Antigens, Ly/metabolism , Encephalitis, California/immunology , Encephalitis, California/metabolism , La Crosse virus , Monocytes/immunology , Monocytes/metabolism , Receptors, CCR2/metabolism , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Brain/immunology , Brain/metabolism , Brain/pathology , Brain/virology , Chemotaxis, Leukocyte/immunology , Disease Models, Animal , Encephalitis, California/virology , Female , Male , Mice , Mice, Transgenic , Monocytes/pathology
17.
Cell Host Microbe ; 22(4): 427-429, 2017 10 11.
Article in English | MEDLINE | ID: mdl-29024636

ABSTRACT

The cellular tropism of noroviruses in immune-competent hosts remains enigmatic. In recent studies in Cell Host & Microbe and Immunity, Lee et al. (2017) and Tomov et al. (2017) determine that norovirus innate immune evasion enables infection of intestinal epithelial cells, escape from adaptive immunity, virus persistence, and shedding.


Subject(s)
Immunity, Innate/immunology , Norovirus , Adaptive Immunity , Epithelial Cells/immunology , Immune Evasion
18.
PLoS One ; 12(3): e0173528, 2017.
Article in English | MEDLINE | ID: mdl-28278296

ABSTRACT

Published data show that murine bone marrow-derived macrophages (BMDM) restrict growth of avirulent phase II, but not virulent phase I, Coxiella burnetii. Growth restriction of phase II bacteria is thought to result from potentiated recognition of pathogen-associated molecular patterns, which leads to production of inhibitory effector molecules. Past studies have used conditioned medium from L-929 murine fibroblasts as a source of macrophage-colony stimulating factor (M-CSF) to promote differentiation of bone marrow-derived myeloid precursors into macrophages. However, uncharacterized components of conditioned medium, such as variable amounts of type I interferons, can affect macrophage activation status and their permissiveness for infection. In the current study, we show that the C. burnetii Nine Mile phase II (NMII) strain grows robustly in primary macrophages from C57BL/6J mice when bone marrow cells are differentiated with recombinant murine M-CSF (rmM-CSF). Bacteria were readily internalized by BMDM, and replicated within degradative, LAMP1-positive vacuoles to achieve roughly 3 logs of growth over 6 days. Uninfected BMDM did not appreciably express CD38 or Egr2, markers of classically (M1) and alternatively (M2) activated macrophages, respectively, nor did infection change the lack of polarization. In accordance with an M0 phenotype, infected BMDM produced moderate amounts of TNF and nitric oxide. Similar NMII growth results were obtained using C57BL/6J myeloid progenitors immortalized with an estrogen-regulated Hoxb8 (ER-Hoxb8) oncogene. To demonstrate the utility of the ER-Hoxb8 system, myeloid progenitors from natural resistance-associated macrophage protein 1 (Nramp1) C57BL/6J knock-in mice were transduced with ER-Hoxb8, and macrophages were derived from immortalized progenitors using rmM-CSF and infected with NMII. No difference in growth was observed when compared to macrophages from wild type mice, indicating depletion of metal ions by the Nramp1 transporter does not negatively impact NMII growth. Results with NMII were recapitulated in primary macrophages where C57BL/6J Nramp1+ BMDM efficiently killed Salmonella enterica serovar Typhimurium. M-CSF differentiated murine macrophages from bone marrow and conditional ER-Hoxb8 myeloid progenitors will be useful ex vivo models for studying Coxiella-macrophage interactions.


Subject(s)
Bone Marrow/microbiology , Coxiella burnetii/growth & development , Macrophages/microbiology , Q Fever/microbiology , Animals , Bone Marrow/metabolism , Cells, Cultured , Colony-Stimulating Factors/metabolism , Coxiella burnetii/pathogenicity , Female , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Q Fever/metabolism , Q Fever/pathology , Tumor Necrosis Factor-alpha/metabolism
19.
Cell Rep ; 18(3): 816-829, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28099857

ABSTRACT

The unprecedented 2013-2016 outbreak of Ebola virus (EBOV) resulted in over 11,300 human deaths. Host resistance to RNA viruses requires RIG-I-like receptor (RLR) signaling through the adaptor protein, mitochondrial antiviral signaling protein (MAVS), but the role of RLR-MAVS in orchestrating anti-EBOV responses in vivo is not known. Here we apply a systems approach to MAVS-/- mice infected with either wild-type or mouse-adapted EBOV. MAVS controlled EBOV replication through the expression of IFNα, regulation of inflammatory responses in the spleen, and prevention of cell death in the liver, with macrophages implicated as a major cell type influencing host resistance. A dominant role for RLR signaling in macrophages was confirmed following conditional MAVS deletion in LysM+ myeloid cells. These findings reveal tissue-specific MAVS-dependent transcriptional pathways associated with resistance to EBOV, and they demonstrate that EBOV adaptation to cause disease in mice involves changes in two distinct events, RLR-MAVS antagonism and suppression of RLR-independent IFN-I responses.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Ebolavirus/physiology , Hemorrhagic Fever, Ebola/pathology , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Animals , DEAD Box Protein 58/antagonists & inhibitors , DEAD Box Protein 58/metabolism , Disease Models, Animal , Hemorrhagic Fever, Ebola/metabolism , Hemorrhagic Fever, Ebola/mortality , Humans , Interferon Type I/metabolism , Kaplan-Meier Estimate , Liver/metabolism , Liver/pathology , Liver/virology , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/cytology , Myeloid Cells/metabolism , Myeloid Cells/virology , Signal Transduction , Spleen/metabolism , Spleen/pathology , Spleen/virology , Virus Replication
20.
Nat Rev Immunol ; 17(3): 195-207, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28111475

ABSTRACT

Two of the most important contemporary emerging viruses that affect human health in Africa are Ebola virus (EBOV) and Lassa virus (LASV). The 2013-2016 West African outbreak of EBOV was responsible for more than 11,000 deaths, primarily in Guinea, Sierra Leone and Liberia. LASV is constantly emerging in these and surrounding West African countries, with an estimate of more than 500,000 cases of Lassa fever, and approximately 5,000 deaths, annually. Both EBOV and LASV are zoonotic, and human infection often results in a severe haemorrhagic fever in both cases. However, the contribution of specific immune responses to disease differs between EBOV and LASV. This Review examines innate and adaptive immune responses to these viruses with the goal of delineating responses that are associated with protective versus pathogenic outcomes.


Subject(s)
Hemorrhagic Fever, Ebola/immunology , Lassa Fever/immunology , Ebolavirus/immunology , Humans , Lassa virus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...