Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Elife ; 102021 07 05.
Article in English | MEDLINE | ID: mdl-34223819

ABSTRACT

Early events in retrovirus transmission are determined by interactions between incoming viruses and frontline cells near entry sites. Despite their importance for retroviral pathogenesis, very little is known about these events. We developed a bioluminescence imaging (BLI)-guided multiscale imaging approach to study these events in vivo. Engineered murine leukemia reporter viruses allowed us to monitor individual stages of retrovirus life cycle including virus particle flow, virus entry into cells, infection and spread for retroorbital, subcutaneous, and oral routes. BLI permitted temporal tracking of orally administered retroviruses along the gastrointestinal tract as they traversed the lumen through Peyer's patches to reach the draining mesenteric sac. Importantly, capture and acquisition of lymph-, blood-, and milk-borne retroviruses spanning three routes was promoted by a common host factor, the I-type lectin CD169, expressed on sentinel macrophages. These results highlight how retroviruses co-opt the immune surveillance function of tissue-resident sentinel macrophages for establishing infection.


Subject(s)
Retroviridae Infections/diagnostic imaging , Retroviridae Infections/transmission , Retroviridae/physiology , Sialic Acid Binding Ig-like Lectin 1/metabolism , Animals , Disease Models, Animal , Female , Humans , Leukemia Virus, Murine , Life Cycle Stages , Lymph Nodes , Macrophages/virology , Male , Mammary Glands, Human/diagnostic imaging , Mammary Glands, Human/virology , Mice , Retroviridae/genetics , Retroviridae Infections/metabolism , Retroviridae Infections/pathology , Sialic Acid Binding Ig-like Lectin 1/genetics , Spleen/diagnostic imaging , Virion , Virus Internalization
2.
PLoS Pathog ; 15(12): e1008161, 2019 12.
Article in English | MEDLINE | ID: mdl-31805155

ABSTRACT

Non-invasive bioluminescent imaging (NIBLI) of HIV-1 infection dynamics allows for real-time monitoring of viral spread and the localization of infected cell populations in living animals. In this report, we describe full-length replication-competent GFP and Nanoluciferase (Nluc) expressing HIV-1 reporter viruses from two clinical transmitted / founder (T/F) strains: TRJO.c and Q23.BG505. By infecting humanized mice with these HIV-1 T/F reporter viruses, we were able to directly monitor longitudinal viral spread at whole-animal resolution via NIBLI at a sensitivity of as few as 30-50 infected cells. Bioluminescent signal strongly correlated with HIV-1 infection and responded proportionally to virus suppression in vivo in animals treated daily with a combination antiretroviral therapy (cART) regimen. Longitudinal NIBLI following cART withdrawal visualized tissue-sites that harbored virus during infection recrudescence. Notably, we observed rebounding infection in the same lymphoid tissues where infection was first observed prior to ART treatment. Our work demonstrates the utility of our system for studying in vivo viral infection dynamics and identifying infected tissue regions for subsequent analyses.


Subject(s)
Anti-HIV Agents/pharmacology , Disease Models, Animal , HIV Infections/virology , HIV-1/drug effects , Luminescent Measurements/methods , Animals , HIV Infections/drug therapy , Humans , Lymphoid Tissue/drug effects , Lymphoid Tissue/virology , Mice , Virus Replication/drug effects
3.
Annu Rev Virol ; 6(1): 501-524, 2019 09 29.
Article in English | MEDLINE | ID: mdl-31283440

ABSTRACT

Viruses are causative agents for many diseases and infect all living organisms on the planet. Development of effective therapies has relied on our ability to isolate and culture viruses in vitro, allowing mechanistic studies and strategic interventions. While this reductionist approach is necessary, testing the relevance of in vitro findings often takes a very long time. New developments in imaging technologies are transforming our experimental approach where viral pathogenesis can be studied in vivo at multiple spatial and temporal resolutions. Here, we outline a vision of a top-down approach using noninvasive whole-body imaging as a guide for in-depth characterization of key tissues, physiologically relevant cell types, and pathways of spread to elucidate mechanisms of virus spread and pathogenesis. Tool development toward imaging of infectious diseases is expected to transform clinical diagnosis and treatment.


Subject(s)
Diagnostic Imaging/methods , Virus Diseases/virology , Virus Physiological Phenomena , Viruses/pathogenicity , Animals , Disease Models, Animal , Humans , Virus Diseases/diagnostic imaging , Virus Diseases/pathology , Virus Replication , Viruses/genetics
4.
Cell Host Microbe ; 25(1): 87-100.e10, 2019 01 09.
Article in English | MEDLINE | ID: mdl-30595553

ABSTRACT

Lymph- and blood-borne retroviruses exploit CD169/Siglec-1-mediated capture by subcapsular sinus and marginal zone metallophilic macrophages for trans-infection of permissive lymphocytes. However, the impact of CD169-mediated virus capture on retrovirus dissemination and pathogenesis in vivo is unknown. In a murine model of the splenomegaly-inducing retrovirus Friend virus complex (FVC) infection, we find that while CD169 promoted draining lymph node infection, it limited systemic spread to the spleen. At the spleen, CD169-expressing macrophages captured incoming blood-borne retroviruses and limited their spread to the erythroblasts in the red pulp where FVC manifests its pathogenesis. CD169-mediated retroviral capture activated conventional dendritic cells 1 (cDC1s) and promoted cytotoxic CD8+ T cell responses, resulting in efficient clearing of FVC-infected cells. Accordingly, CD169 blockade led to higher viral loads and accelerated death in susceptible mouse strains. Thus, CD169 plays a protective role during FVC pathogenesis by reducing viral dissemination to erythroblasts and eliciting an effective cytotoxic T lymphocyte response via cDC1s.


Subject(s)
Lectins/pharmacology , Protective Agents/pharmacology , Retroviridae Infections/drug therapy , Retroviridae/drug effects , Retroviridae/pathogenicity , Sialic Acid Binding Ig-like Lectin 1/pharmacology , Animals , CD8-Positive T-Lymphocytes , Cell Proliferation , Dendritic Cells/virology , Disease Models, Animal , Erythroblasts/virology , Female , Interferon Type I/metabolism , Lymph Nodes/virology , Macrophages/drug effects , Macrophages/virology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Spleen , T-Lymphocytes, Cytotoxic , Viral Load
5.
Virol J ; 11: 100, 2014 May 28.
Article in English | MEDLINE | ID: mdl-24884573

ABSTRACT

BACKGROUND: The antiviral protein Daxx acts as a restriction factor of avian sarcoma virus (ASV; Retroviridae) in mammalian cells by promoting epigenetic silencing of integrated proviral DNA. Although Daxx is encoded by a type I (α/ß) interferon-stimulated gene, the requirement for Daxx in the interferon anti-retroviral response has not been elucidated. In this report, we describe the results of experiments designed to investigate the role of Daxx in the type I interferon-induced anti-ASV response. FINDINGS: Using an ASV reporter system, we show that type I interferons are potent inhibitors of ASV replication. We demonstrate that, while Daxx is necessary to silence ASV gene expression in the absence of interferons, type I interferons are fully-capable of inducing an antiviral state in the absence of Daxx. CONCLUSIONS: These results provide evidence that Daxx is not essential for the anti-ASV interferon response in mammalian cells, and that interferons deploy multiple, redundant antiviral mechanisms to protect cells from ASV.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Avian Sarcoma Viruses/immunology , Avian Sarcoma Viruses/physiology , Interferon Type I/immunology , Nuclear Proteins/immunology , Virus Replication , Animals , Birds , Cell Line , Co-Repressor Proteins , Humans , Molecular Chaperones
SELECTION OF CITATIONS
SEARCH DETAIL
...