Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
1.
Cell ; 185(13): 2354-2369.e17, 2022 06 23.
Article in English | MEDLINE | ID: mdl-35568036

ABSTRACT

Interferons (IFNs) induce an antimicrobial state, protecting tissues from infection. Many viruses inhibit IFN signaling, but whether bacterial pathogens evade IFN responses remains unclear. Here, we demonstrate that the Shigella OspC family of type-III-secreted effectors blocks IFN signaling independently of its cell death inhibitory activity. Rather, IFN inhibition was mediated by the binding of OspC1 and OspC3 to the Ca2+ sensor calmodulin (CaM), blocking CaM kinase II and downstream JAK/STAT signaling. The growth of Shigella lacking OspC1 and OspC3 was attenuated in epithelial cells and in a murine model of infection. This phenotype was rescued in both models by the depletion of IFN receptors. OspC homologs conserved in additional pathogens not only bound CaM but also inhibited IFN, suggesting a widespread virulence strategy. These findings reveal a conserved but previously undescribed molecular mechanism of IFN inhibition and demonstrate the critical role of Ca2+ and IFN targeting in bacterial pathogenesis.


Subject(s)
Interferons , Virulence Factors , Animals , Antiviral Agents , Calcium Signaling , Epithelial Cells/metabolism , Interferons/metabolism , Mice , Virulence Factors/metabolism
2.
Cell ; 182(2): 515-530.e17, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32610083

ABSTRACT

Imaging of biological matter across resolution scales entails the challenge of preserving the direct and unambiguous correlation of subject features from the macroscopic to the microscopic level. Here, we present a correlative imaging platform developed specifically for imaging cells in 3D under cryogenic conditions by using X-rays and visible light. Rapid cryo-preservation of biological specimens is the current gold standard in sample preparation for ultrastructural analysis in X-ray imaging. However, cryogenic fluorescence localization methods are, in their majority, diffraction-limited and fail to deliver matching resolution. We addressed this technological gap by developing an integrated, user-friendly platform for 3D correlative imaging of cells in vitreous ice by using super-resolution structured illumination microscopy in conjunction with soft X-ray tomography. The power of this approach is demonstrated by studying the process of reovirus release from intracellular vesicles during the early stages of infection and identifying intracellular virus-induced structures.


Subject(s)
Cryoelectron Microscopy/methods , Reoviridae/physiology , Cell Line, Tumor , Cryoelectron Microscopy/instrumentation , Endosomes/metabolism , Endosomes/virology , Fluorescent Dyes/chemistry , Humans , Imaging, Three-Dimensional , Microscopy, Fluorescence , Reoviridae/chemistry , Virus Release/physiology
3.
Cell ; 150(3): 495-507, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22863004

ABSTRACT

Coated pits assemble by growth of a clathrin lattice, which is linked by adaptors to the underlying membrane. How does this process start? We used live-cell TIRF imaging with single-molecule EGFP sensitivity and high temporal resolution to detect arrival of the clathrin triskelions and AP2 adaptors that initiate coat assembly. Unbiased object identification and trajectory tracking, together with a statistical model, yield the arrival times and numbers of individual proteins, as well as experimentally confirmed estimates of the extent of substitution of endogenous by expressed, fluorescently tagged proteins. Pits initiate by coordinated arrival of clathrin and AP2, which is usually detected as two sequential steps, each of one triskelion with two adaptors. PI-4,5-P2 is essential for initiation. The accessory proteins FCHo1/2 are not; instead, they are required for sustained growth. This objective picture of coated pit initiation also shows that methods outlined here will be broadly useful for studies of dynamic assemblies in living cells.


Subject(s)
Clathrin/metabolism , Coated Pits, Cell-Membrane/metabolism , Membrane Proteins/metabolism , Adaptor Protein Complex 2/metabolism , Animals , Cell Line , Cell Membrane/metabolism , Chlorocebus aethiops , Proteins/metabolism
4.
Nat Immunol ; 15(8): 717-26, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24952503

ABSTRACT

Type I interferon responses are considered the primary means by which viral infections are controlled in mammals. Despite this view, several pathogens activate antiviral responses in the absence of type I interferons. The mechanisms controlling type I interferon-independent responses are undefined. We found that RIG-I like receptors (RLRs) induce type III interferon expression in a variety of human cell types, and identified factors that differentially regulate expression of type I and type III interferons. We identified peroxisomes as a primary site of initiation of type III interferon expression, and revealed that the process of intestinal epithelial cell differentiation upregulates peroxisome biogenesis and promotes robust type III interferon responses in human cells. These findings highlight the importance of different intracellular organelles in specific innate immune responses.


Subject(s)
Immunity, Innate , Interferons/immunology , Peroxisomes/immunology , Animals , Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Cell Differentiation , Cell Line , Cyclohexanes/pharmacology , DEAD Box Protein 58 , DEAD-box RNA Helicases/immunology , Enzyme Inhibitors/pharmacology , Humans , Interferons/biosynthesis , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/genetics , Mice , Pyridones/pharmacology , RNA Interference , RNA, Small Interfering , Receptors, Immunologic , Reoviridae/immunology , Reoviridae Infections/immunology , STAT1 Transcription Factor/antagonists & inhibitors , STAT1 Transcription Factor/immunology , Signal Transduction/immunology , Tyrphostins/pharmacology , Vidarabine/analogs & derivatives , Vidarabine/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics
5.
EMBO J ; 40(16): e107821, 2021 08 16.
Article in English | MEDLINE | ID: mdl-34159616

ABSTRACT

SARS-CoV-2 is a newly emerged coronavirus that caused the global COVID-19 outbreak in early 2020. COVID-19 is primarily associated with lung injury, but many other clinical symptoms such as loss of smell and taste demonstrated broad tissue tropism of the virus. Early SARS-CoV-2-host cell interactions and entry mechanisms remain poorly understood. Investigating SARS-CoV-2 infection in tissue culture, we found that the protease TMPRSS2 determines the entry pathway used by the virus. In the presence of TMPRSS2, the proteolytic process of SARS-CoV-2 was completed at the plasma membrane, and the virus rapidly entered the cells within 10 min in a pH-independent manner. When target cells lacked TMPRSS2 expression, the virus was endocytosed and sorted into endolysosomes, from which SARS-CoV-2 entered the cytosol via acid-activated cathepsin L protease 40-60 min post-infection. Overexpression of TMPRSS2 in non-TMPRSS2 expressing cells abolished the dependence of infection on the cathepsin L pathway and restored sensitivity to the TMPRSS2 inhibitors. Together, our results indicate that SARS-CoV-2 infects cells through distinct, mutually exclusive entry routes and highlight the importance of TMPRSS2 for SARS-CoV-2 sorting into either pathway.


Subject(s)
COVID-19/metabolism , Cathepsin L/metabolism , SARS-CoV-2/physiology , Serine Endopeptidases/metabolism , Animals , COVID-19/genetics , Caco-2 Cells , Chlorocebus aethiops , Endocytosis , Host Microbial Interactions , Humans , Hydrogen-Ion Concentration , Proteolysis , Serine Endopeptidases/genetics , Signal Transduction , Vero Cells , Virus Internalization
6.
Mol Syst Biol ; 20(3): 242-275, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38273161

ABSTRACT

Isogenic cells respond in a heterogeneous manner to interferon. Using a micropatterning approach combined with high-content imaging and spatial analyses, we characterized how the population context (position of a cell with respect to neighboring cells) of epithelial cells affects their response to interferons. We identified that cells at the edge of cellular colonies are more responsive than cells embedded within colonies. We determined that this spatial heterogeneity in interferon response resulted from the polarized basolateral interferon receptor distribution, making cells located in the center of cellular colonies less responsive to ectopic interferon stimulation. This was conserved across cell lines and primary cells originating from epithelial tissues. Importantly, cells embedded within cellular colonies were not protected from viral infection by apical interferon treatment, demonstrating that the population context-driven heterogeneous response to interferon influences the outcome of viral infection. Our data highlights that the behavior of isolated cells does not directly translate to their behavior in a population, placing the population context as one important factor influencing heterogeneity during interferon response in epithelial cells.


Subject(s)
Interferons , Virus Diseases , Humans , Interferons/pharmacology , Interferons/metabolism , Epithelial Cells/metabolism , Cell Line , Virus Diseases/metabolism
7.
Cell ; 141(4): 668-81, 2010 May 14.
Article in English | MEDLINE | ID: mdl-20451243

ABSTRACT

Peroxisomes have long been established to play a central role in regulating various metabolic activities in mammalian cells. These organelles act in concert with mitochondria to control the metabolism of lipids and reactive oxygen species. However, while mitochondria have emerged as an important site of antiviral signal transduction, a role for peroxisomes in immune defense is unknown. Here, we report that the RIG-I-like receptor (RLR) adaptor protein MAVS is located on peroxisomes and mitochondria. We find that peroxisomal and mitochondrial MAVS act sequentially to create an antiviral cellular state. Upon viral infection, peroxisomal MAVS induces the rapid interferon-independent expression of defense factors that provide short-term protection, whereas mitochondrial MAVS activates an interferon-dependent signaling pathway with delayed kinetics, which amplifies and stabilizes the antiviral response. The interferon regulatory factor IRF1 plays a crucial role in regulating MAVS-dependent signaling from peroxisomes. These results establish that peroxisomes are an important site of antiviral signal transduction.


Subject(s)
Immunity, Innate , Peroxisomes/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line , Chlorocebus aethiops , Fibroblasts/metabolism , Hepatocytes/metabolism , Humans , Interferons/metabolism , Mice , Mitochondria/metabolism , Vero Cells
8.
Cell Mol Life Sci ; 81(1): 71, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38300320

ABSTRACT

Hexosylceramides (HexCer) are implicated in the infection process of various pathogens. However, the molecular and cellular functions of HexCer in infectious cycles are poorly understood. Investigating the enveloped virus Uukuniemi (UUKV), a bunyavirus of the Phenuiviridae family, we performed a lipidomic analysis with mass spectrometry and determined the lipidome of both infected cells and derived virions. We found that UUKV alters the processing of HexCer to glycosphingolipids (GSL) in infected cells. The infection resulted in the overexpression of glucosylceramide (GlcCer) synthase (UGCG) and the specific accumulation of GlcCer and its subsequent incorporation into viral progeny. UUKV and several pathogenic bunyaviruses relied on GlcCer in the viral envelope for binding to various host cell types. Overall, our results indicate that GlcCer is a structural determinant of virions crucial for bunyavirus infectivity. This study also highlights the importance of glycolipids on virions in facilitating interactions with host cell receptors and infectious entry of enveloped viruses.


Subject(s)
Orthobunyavirus , Glucosylceramides , Virus Attachment , Lipidomics , Mass Spectrometry
9.
J Virol ; 97(11): e0097223, 2023 Nov 30.
Article in English | MEDLINE | ID: mdl-37909728

ABSTRACT

IMPORTANCE: The current view is that the default pathway of Kaposi's sarcoma-associated herpesvirus (KSHV) infection is the establishment of latency, which is a prerequisite for lifelong infection and viral oncogenesis. This view about KSHV infection is supported by the observations that KSHV latently infects most of the cell lines cultured in vitro in the absence of any environmental stresses that may occur in vivo. The goal of this study was to determine the effect of hypoxia, a natural stress stimulus, on primary KSHV infection. Our data indicate that hypoxia promotes euchromatin formation on the KSHV genome following infection and supports lytic de novo KSHV infection. We also discovered that hypoxia-inducible factor-1α is required and sufficient for allowing lytic KSHV infection. Based on our results, we propose that hypoxia promotes lytic de novo infection in cells that otherwise support latent infection under normoxia; that is, the environmental conditions can determine the outcome of KSHV primary infection.


Subject(s)
Herpesviridae Infections , Hypoxia-Inducible Factor 1, alpha Subunit , Hypoxia , Humans , Gene Expression Regulation, Viral , Herpesvirus 8, Human , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Sarcoma, Kaposi , Virus Latency
10.
J Virol ; 97(2): e0008923, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36700640

ABSTRACT

Viruses have brought humanity many challenges: respiratory infection, cancer, neurological impairment and immunosuppression to name a few. Virology research over the last 60+ years has responded to reduce this disease burden with vaccines and antivirals. Despite this long history, the COVID-19 pandemic has brought unprecedented attention to the field of virology. Some of this attention is focused on concern about the safe conduct of research with human pathogens. A small but vocal group of individuals has seized upon these concerns - conflating legitimate questions about safely conducting virus-related research with uncertainties over the origins of SARS-CoV-2. The result has fueled public confusion and, in many instances, ill-informed condemnation of virology. With this article, we seek to promote a return to rational discourse. We explain the use of gain-of-function approaches in science, discuss the possible origins of SARS-CoV-2 and outline current regulatory structures that provide oversight for virological research in the United States. By offering our expertise, we - a broad group of working virologists - seek to aid policy makers in navigating these controversial issues. Balanced, evidence-based discourse is essential to addressing public concern while maintaining and expanding much-needed research in virology.


Subject(s)
Research , Virology , Virus Diseases , Humans , COVID-19/prevention & control , Information Dissemination , Pandemics/prevention & control , Policy Making , Research/standards , Research/trends , SARS-CoV-2 , Virology/standards , Virology/trends , Virus Diseases/prevention & control , Virus Diseases/virology , Viruses
SELECTION OF CITATIONS
SEARCH DETAIL