Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Sci Signal ; 16(803): eade3599, 2023 09 19.
Article in English | MEDLINE | ID: mdl-37725663

ABSTRACT

RUBCN (also known as Rubicon) was originally identified as a negative regulator of autophagy, a process by which cells degrade and recycle damaged components or organelles and that requires the activity of the class III PI3K VPS34 and the mTORC1 protein complex. Here, we characterized the role of a shorter isoform, RUBCN100, as an autophagy-promoting factor in B cells. RUBCN100 was translated from alternative translation initiation sites and lacked the RUN domain of the longer, previously characterized RUBCN130 isoform. Specific deficiency of RUBCN130 in B cells enhanced autophagy, which promoted memory B cell generation. In contrast to RUBCN130, which is localized in late endosomes and lysosomes and suppresses the enzymatic activity of VPS34, an effect thought to mediated by its RUN domain, RUBCN100 was preferentially located in early endosomes and enhanced VPS34 activity, presumably because of the absence of the RUN domain. Furthermore, RUBCN100, but not RUBCN130, enhanced autophagy and suppressed mTORC1 activation. Our findings reveal that the opposing roles of two RUBCN isoforms are critical for autophagy regulation and memory B cell differentiation.


Subject(s)
B-Lymphocytes , Memory B Cells , Autophagy , Protein Isoforms/genetics , Mechanistic Target of Rapamycin Complex 1/genetics
2.
Sci Rep ; 13(1): 11610, 2023 07 18.
Article in English | MEDLINE | ID: mdl-37463955

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes severe acute respiratory symptoms in humans. Controlling the coronavirus disease pandemic is a worldwide priority. The number of SARS-CoV-2 studies has dramatically increased, and the requirement for analytical tools is higher than ever. Here, we propose monolayered-intestinal epithelial cells (IECs) derived from human induced pluripotent stem cells (iPSCs) instead of three-dimensional cultured intestinal organoids as a suitable tool to study SARS-CoV-2 infection. Differentiated IEC monolayers express high levels of angiotensin-converting enzyme 2 and transmembrane protease serine 2 (TMPRSS2), host factors essential for SARS-CoV-2 infection. SARS-CoV-2 efficiently grows in IEC monolayers. Using this propagation system, we confirm that TMPRSS2 inhibition blocked SARS-CoV-2 infection in IECs. Hence, our iPSC-derived IEC monolayers are suitable for SARS-CoV-2 research under physiologically relevant conditions.


Subject(s)
COVID-19 , Induced Pluripotent Stem Cells , Humans , SARS-CoV-2 , Epithelial Cells , Intestines
3.
Lab Chip ; 23(4): 609-623, 2023 02 14.
Article in English | MEDLINE | ID: mdl-36633172

ABSTRACT

Three-dimensional (3D) cell culture, which provides an in vivo-like environment in vitro unlike the conventional two-dimensional (2D) cell culture, has attracted much attention from researchers. Although various 3D cell culture methods have been developed, information on a method using inorganic nanoclay is scant. Here, we report that hectorite, an inorganic layered silicate, can be used as an auxiliary material for 3D cell culture. Human colon cancer cell lines cultured in a medium containing 0.01% synthetic hectorite spontaneously formed 3D spheroids in an adherent plate. Morphologically, these spheroids were more dispersed in all directions than control spheroids generated in an ultralow adherent plate. Microarray analysis showed that FGF19, TGM2, and SERPINA3, whose expression is reportedly increased in colon cancer tissues and is related to tumorigenesis or metastasis, were upregulated in HT-29 spheroids formed using synthetic hectorite compared with those in control spheroids. Gene ontology analysis revealed upregulation of genes associated with morphogenesis, cytoskeleton, extracellular matrix, cellular uptake and secretion, signaling pathways, and gene expression regulation. Moreover, fluorescence-labeled hectorite particles were localized in the cytoplasm of individual cells in spheroids. These results suggest that the synthetic hectorite modified the physiological state of and gene expression within the cells, triggering spheroid formation with malignant characteristics. Our findings highlight a novel application of synthetic hectorite for 3D cell culture.


Subject(s)
Colorectal Neoplasms , Spheroids, Cellular , Humans , Cell Culture Techniques/methods , Silicates/pharmacology
4.
mBio ; 13(4): e0123322, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35862783

ABSTRACT

Group A Streptococcus (GAS), a deleterious human-pathogenic bacterium, causes life-threatening diseases such as sepsis and necrotic fasciitis. We recently reported that GAS survives and replicates within blood vessel endothelial cells because these cells are intrinsically defective in xenophagy. Because blood vessel endothelial cells are relatively germfree environments, specific stimulation may be required to sufficiently induce xenophagy. Here, we explored how vascular endothelial growth factor (VEGF) promoted xenophagy and lysosomal activity in endothelial cells. These effects were achieved by amplifying the activation of TFEB, a transcriptional factor crucial for lysosome/autophagy biogenesis, via cAMP-mediated calcium release. In a mouse model of local infection with GAS, the VEGF level was significantly elevated at the infection site. Interestingly, low serum VEGF levels were found in a mouse model of invasive bacteremia and in patients with severe GAS-induced sepsis. Moreover, the administration of VEGF improved the survival of GAS-infected mice. We propose a novel theory regarding GAS infection in endothelial cells, wherein VEGF concentrations in the systemic circulation play a critical role. IMPORTANCE Sepsis caused by Streptococcus pyogenes is a life-threatening condition. Blood vessel endothelial cells should serve as a barrier to infection, although we recently reported that endothelial cells allow intracellular GAS proliferation due to defective xenophagy. In this study, we revealed that administration of VEGF augmented both xenophagy and lysosomal activity in these cells, leading to the efficient killing of intracellular GAS. By comparison, the opposite relationship was observed in vivo, as low serum VEGF concentrations were accompanied by high-severity sepsis in both a mouse model and in human patients. Administration of VEGF reduced mortality in the GAS sepsis model. Based on these findings, we hypothesize that during acute infection, strong VEGF stimulation boosts the intracellular defense system of the endothelium to provide a stronger blood vessel barrier, thereby helping to prevent bacterial dissemination.


Subject(s)
Sepsis , Streptococcus pyogenes , Animals , Autophagy , Endothelial Cells/microbiology , Humans , Lysosomes , Mice , Streptococcus pyogenes/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factors/metabolism
7.
J Cell Biol ; 219(8)2020 08 03.
Article in English | MEDLINE | ID: mdl-32492081

ABSTRACT

In macroautophagy, membrane structures called autophagosomes engulf substrates and deliver them for lysosomal degradation. Autophagosomes enwrap a variety of targets with diverse sizes, from portions of cytosol to larger organelles. However, the mechanism by which autophagosome size is controlled remains elusive. We characterized a novel ER membrane protein, ERdj8, in mammalian cells. ERdj8 localizes to a meshwork-like ER subdomain along with phosphatidylinositol synthase (PIS) and autophagy-related (Atg) proteins. ERdj8 overexpression extended the size of the autophagosome through its DnaJ and TRX domains. ERdj8 ablation resulted in a defect in engulfing larger targets. C. elegans, in which the ERdj8 orthologue dnj-8 was knocked down, could perform autophagy on smaller mitochondria derived from the paternal lineage but not the somatic mitochondria. Thus, ERdj8 may play a critical role in autophagosome formation by providing the capacity to target substrates of diverse sizes for degradation.


Subject(s)
Autophagosomes/metabolism , Endoplasmic Reticulum/metabolism , HSP40 Heat-Shock Proteins/metabolism , Macroautophagy , Animals , Animals, Genetically Modified , Autophagosomes/genetics , Autophagosomes/ultrastructure , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , CDP-Diacylglycerol-Inositol 3-Phosphatidyltransferase/genetics , CDP-Diacylglycerol-Inositol 3-Phosphatidyltransferase/metabolism , COS Cells , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Chlorocebus aethiops , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/ultrastructure , HSP40 Heat-Shock Proteins/genetics , HeLa Cells , Humans , Mitochondria/metabolism , Mitochondria/ultrastructure
8.
Front Microbiol ; 11: 117, 2020.
Article in English | MEDLINE | ID: mdl-32117141

ABSTRACT

Group A streptococcus (GAS) is a versatile pathogen that causes a wide spectrum of diseases in humans. Invading host cells is a known strategy for GAS to avoid antibiotic killing and immune recognition. However, the underlying mechanisms of GAS resistance to intracellular killing need to be explored. Endothelial HMEC-1 cells were infected with GAS, methicillin-resistant Staphylococcus aureus (MRSA) and Salmonella Typhimurium under nicotinamide (NAM)-supplemented conditions. The intracellular NAD+ level and cell viability were respectively measured by NAD+ quantification kit and protease-based cytotoxicity assay. Moreover, the intracellular bacteria were analyzed by colony-forming assay, transmission electron microscopy, and confocal microscopy. We found that supplementation with exogenous nicotinamide during infection significantly inhibited the growth of intracellular GAS in endothelial cells. Moreover, the NAD+ content and NAD+/NADH ratio of GAS-infected endothelial cells were dramatically increased, whereas the cell cytotoxicity was decreased by exogenous nicotinamide treatment. After knockdown of the autophagy-related ATG9A, the intracellular bacterial load was increased in nicotinamide-treated endothelial cells. The results of Western blot and transmission electron microscopy also revealed that cells treated with nicotinamide can increase autophagy-associated LC3 conversion and double-membrane formation during GAS infection. Confocal microscopy images further showed that more GAS-containing vacuoles were colocalized with lysosome under nicotinamide-supplemented conditions than without nicotinamide treatment. In contrast to GAS, supplementation with exogenous nicotinamide did not effectively inhibit the growth of MRSA or S. Typhimurium in endothelial cells. These results indicate that intracellular NAD+ homeostasis is crucial for controlling intracellular GAS infection in endothelial cells. In addition, nicotinamide may be a potential new therapeutic agent to overcome persistent infections of GAS.

10.
mBio ; 10(5)2019 10 01.
Article in English | MEDLINE | ID: mdl-31575768

ABSTRACT

Group A streptococcus (GAS) is an important human pathogen which can cause fatal diseases after invasion into the bloodstream. Although antibiotics and immune surveillance are the main defenses against GAS infection, GAS utilizes internalization into cells as a major immune evasion strategy. Our previous findings revealed that light chain 3 (LC3)-associated single membrane GAS-containing vacuoles in endothelial cells are compromised for bacterial clearance due to insufficient acidification after fusion with lysosomes. However, the characteristics and the activation mechanisms of these LC3-positive compartments are still largely unknown. In the present study, we demonstrated that the LC3-positive GAS is surrounded by single membrane and colocalizes with NADPH oxidase 2 (NOX2) complex but without ULK1, which are characteristics of LC3-associated phagocytosis (LAP). Inhibition of NOX2 or reactive oxygen species (ROS) significantly reduces GAS multiplication and enhances autolysosome acidification in endothelial cells through converting LAP to conventional xenophagy, which is revealed by enhancement of ULK1 recruitment, attenuation of p70s6k phosphorylation, and formation of the isolation membrane. We also clarify that the inactivation of mTORC1, which is the initiation signal of autophagy, is inhibited by NOX2- and ROS-activated phosphatidylinositol 3-kinase (PI3K)/AKT and MEK/extracellular signal-regulated kinase (ERK) pathways. In addition, streptolysin O (SLO) of GAS is identified as a crucial inducer of ROS for ß1 integrin-mediated LAP induction. After downregulation of ß1 integrin, GAS multiplication is reduced, accompanied with LAP inhibition and xenophagy induction. These results demonstrate that GAS infection preferentially induces ineffective LAP to evade xenophagic killing in endothelial cells through the SLO/ß1 integrin/NOX2/ROS pathway.IMPORTANCE Our previous reports showed that the LC3-associated GAS-containing single membrane vacuoles are inefficient for bacterial clearance in endothelial cells, which may result in bacteremia. However, the characteristics and the induction mechanisms of these LC3-positive vacuoles are still largely unknown. Here we provide the first evidence that these LC3-positive GAS-containing single membrane compartments appear to be LAPosomes, which are induced by NOX2 and ROS. Through NOX2- and ROS-mediated signaling, GAS preferentially induces LAP and inhibits bacteriostatic xenophagy in endothelial cells. We also provide the first demonstration that ß1 integrin acts as the receptor for LAP induction through GAS-produced SLO stimulation in endothelial cells. Our findings reveal the underlying mechanisms of LAP induction and autophagy evasion for GAS multiplication in endothelial cells.


Subject(s)
Endothelial Cells/microbiology , Macroautophagy , Streptococcus pyogenes/physiology , Streptolysins/metabolism , Bacterial Proteins/metabolism , Cell Line , Humans , Integrin beta1/metabolism , Microtubule-Associated Proteins/metabolism , NADPH Oxidase 2/metabolism , Reactive Oxygen Species/metabolism , Vacuoles/metabolism
11.
Virus Res ; 272: 197732, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31445103

ABSTRACT

To visually examine the early phase of chikungunya virus (CHIKV) infection in target cells, we constructed a virus-like particle (VLP) in which the envelope protein E1 is fused with green fluorescent protein (GFP). This chikungunya VLP-GFP (CHIK-VLP-EGFP), purified by density gradient fractionation, was observed as 60-70 nm-dia. particles and was detected as tiny puncta of fluorescence in the cells. CHIK-VLP-EGFP showed binding properties similar to those of the wild-type viruses. Most of the fluorescence signals that had bound on Vero cells disappeared within 30 min at 37 °C, but not in the presence of anti-CHIKV neutralizing serum or an endosomal acidification inhibitor (bafilomycin A1), suggesting that the loss of fluorescence signals is due to the disassembly of the viral envelope following the internalization of CHIK-VLP-EGFP. In addition to these results, the fluorescence signals disappeared in highly susceptible Vero and U251MG cells but not in poorly susceptible A549 cells. Thus, CHIK-VLP-EGFP is a useful tool to examine the effects of the CHIKV neutralizing antibodies and antiviral compounds that are effective in the entry phase of CHIKV.


Subject(s)
Chikungunya Fever/virology , Chikungunya virus/physiology , Genes, Reporter , Green Fluorescent Proteins/genetics , Virus Replication , Animals , Cells, Cultured , Chikungunya virus/ultrastructure , Chlorocebus aethiops , Gene Expression , Genetic Vectors/genetics , Models, Biological , Vero Cells , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Virus Internalization
12.
Methods Mol Biol ; 1998: 73-92, 2019.
Article in English | MEDLINE | ID: mdl-31250295

ABSTRACT

Many enveloped viruses utilize the cellular ESCRT pathway for budding, even flaviviruses, which form viral particles inside replication organelles derived from the endoplasmic reticulum (ER). In this section, we introduce methods for detecting several ESCRT subunit proteins in virus-infected cells by immunofluorescence microscopy and immunoelectron microscopy (immuno-EM). We also introduce a new method; correlative light microscopy and electron microscopy (CLEM), which allows the observation of target structures with both high-resolution EM and fluorescence labeling.


Subject(s)
Biological Assay/methods , Endoplasmic Reticulum/ultrastructure , Endosomal Sorting Complexes Required for Transport/metabolism , Microscopy, Electron, Transmission/methods , Molecular Imaging/methods , Animals , Cell Culture Techniques/methods , Cell Line , Dengue Virus/immunology , Encephalitis Virus, Japanese/immunology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/virology , Endosomal Sorting Complexes Required for Transport/chemistry , Endosomal Sorting Complexes Required for Transport/immunology , Gold/chemistry , Humans , Mesocricetus , Metal Nanoparticles/chemistry , Microscopy, Fluorescence/methods , Microscopy, Immunoelectron/methods , Staining and Labeling/methods , Virion/chemistry
13.
Stem Cell Res ; 23: 50-56, 2017 08.
Article in English | MEDLINE | ID: mdl-28689068

ABSTRACT

We sought to establish a more efficient technique for induction of inner ear hair cell-like cells (HC-like cells) from embryonic stem cells (ES cells) by using a combination of two previously reported methods; ST2 stromal cell-conditioned medium, known to be favorable for HC-like cell induction (HIST2 method), and ES cells with transfer of the Math1 gene (Math1-ES cells). Math1-ES cells carrying Tet-inducible Math1 were cultured for 14days with doxycycline in conditioned medium from cultures of ST2 stromal cells following formation of 4-day embryoid bodies (EBs). Although each of the previously introduced methods have been reported to induce approximately 20% HC-like cells and 10% HC-like cells in their respective populations in EB outgrowths at the end of the culture periods, the present combined method was able to generate approximately 30% HC-like cells expressing HC-related markers (myosin6, myosin7a, calretinin, α9AchR, Brn3c), which showed remarkable formation of stereocilia-like structures. Analysis of expressions of marker genes specific for cochlear (Lmod3, Emcn) and vestibular (Dnah5, Ptgds) cells indicated that our HIST2 method may lead to induction of cochlear- and vestibular-type cells. In addition, continuous Math1 induction by doxycycline without use of the HIST2 method preferentially induced cochlear markers with negligible effects on vestibular marker induction.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Culture Media, Conditioned/pharmacology , Hair Cells, Auditory, Inner/cytology , Mouse Embryonic Stem Cells/cytology , Transfection , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers/metabolism , Cell Line , Cells, Cultured , Cochlea/cytology , Embryoid Bodies/cytology , Embryoid Bodies/drug effects , Gene Expression Regulation/drug effects , Mechanotransduction, Cellular , Mice , Myosins/metabolism , Stereocilia/metabolism , Stromal Cells/metabolism , Vestibule, Labyrinth/cytology
14.
PLoS Pathog ; 13(7): e1006444, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28683091

ABSTRACT

Group A Streptococcus (GAS) is deleterious pathogenic bacteria whose interaction with blood vessels leads to life-threatening bacteremia. Although xenophagy, a special form of autophagy, eliminates invading GAS in epithelial cells, we found that GAS could survive and multiply in endothelial cells. Endothelial cells were competent in starvation-induced autophagy, but failed to form double-membrane structures surrounding GAS, an essential step in xenophagy. This deficiency stemmed from reduced recruitment of ubiquitin and several core autophagy proteins in endothelial cells, as demonstrated by the fact that it could be rescued by exogenous coating of GAS with ubiquitin. The defect was associated with reduced NO-mediated ubiquitin signaling. Therefore, we propose that the lack of efficient clearance of GAS in endothelial cells is caused by their intrinsic inability to target GAS with ubiquitin to promote autophagosome biogenesis for xenophagy.


Subject(s)
Autophagy , Endothelial Cells/cytology , Streptococcal Infections/physiopathology , Streptococcus pyogenes/physiology , Cell Line , Endothelial Cells/metabolism , Endothelial Cells/microbiology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Host-Pathogen Interactions , Humans , Phagosomes/metabolism , Phagosomes/microbiology , Streptococcal Infections/metabolism , Streptococcal Infections/microbiology , Streptococcus pyogenes/genetics , Ubiquitin/metabolism
15.
Virology ; 510: 165-174, 2017 10.
Article in English | MEDLINE | ID: mdl-28738245

ABSTRACT

Infection with coronavirus rearranges the host cell membrane to assemble a replication/transcription complex in which replication of the viral genome and transcription of viral mRNA occur. Although coexistence of nsp3 and nsp4 is known to cause membrane rearrangement, the mechanisms underlying the interaction of these two proteins remain unclear. We demonstrated that binding of nsp4 with nsp3 is essential for membrane rearrangement and identified amino acid residues in nsp4 responsible for the interaction with nsp3. In addition, we revealed that the nsp3-nsp4 interaction is not sufficient to induce membrane rearrangement, suggesting the participation of other factors such as host proteins. Finally, we showed that loss of the nsp3-nsp4 interaction eliminated viral replication by using an infectious cDNA clone and replicon system of SARS-CoV. These findings provide clues to the mechanism of the replication/transcription complex assembly of SARS-CoV and could reveal an antiviral target for the treatment of betacoronavirus infection.


Subject(s)
Amino Acid Substitution , Severe acute respiratory syndrome-related coronavirus/physiology , Viral Nonstructural Proteins/genetics , Virus Replication , DNA Mutational Analysis , Protein Interaction Mapping , Severe acute respiratory syndrome-related coronavirus/genetics
16.
Cell Rep ; 16(9): 2339-47, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27545892

ABSTRACT

Flavivirus infection induces endoplasmic reticulum (ER) membrane rearrangements to generate a compartment for replication of the viral genome and assembly of viral particles. Using quantitative mass spectrometry, we identified several ESCRT (endosomal sorting complex required for transport) proteins that are recruited to sites of virus replication on the ER. Systematic small interfering RNA (siRNA) screening revealed that release of both dengue virus and Japanese encephalitis virus was dramatically decreased by single depletion of TSG101 or co-depletion of specific combinations of ESCRT-III proteins, resulting in ≥1,000-fold titer reductions. By contrast, release was unaffected by depletion of some core ESCRTs, including VPS4. Reintroduction of ESCRT proteins to siRNA-depleted cells revealed interactions among ESCRT proteins that are crucial for flavivirus budding. Electron-microscopy studies revealed that the CHMP2 and CHMP4 proteins function directly in membrane deformation at the ER. Thus, a unique and specific subset of ESCRT contributes to ER membrane biogenesis during flavivirus infection.


Subject(s)
DNA-Binding Proteins/genetics , Dengue Virus/genetics , Encephalitis Virus, Japanese/genetics , Endoplasmic Reticulum/genetics , Endosomal Sorting Complexes Required for Transport/genetics , Epithelial Cells/metabolism , Transcription Factors/genetics , Virion/genetics , ATPases Associated with Diverse Cellular Activities , Animals , Cell Line , Chlorocebus aethiops , Cricetulus , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Dengue Virus/growth & development , Dengue Virus/metabolism , Encephalitis Virus, Japanese/growth & development , Encephalitis Virus, Japanese/metabolism , Endoplasmic Reticulum/virology , Endosomal Sorting Complexes Required for Transport/antagonists & inhibitors , Endosomal Sorting Complexes Required for Transport/metabolism , Epithelial Cells/virology , Gene Expression Regulation , HEK293 Cells , Host-Pathogen Interactions , Humans , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , Vacuolar Proton-Translocating ATPases/genetics , Vacuolar Proton-Translocating ATPases/metabolism , Vero Cells , Virion/metabolism , Virus Replication/genetics
17.
Virology ; 496: 51-58, 2016 09.
Article in English | MEDLINE | ID: mdl-27254595

ABSTRACT

In latent infection of Kaposi's sarcoma-associated herpesvirus (KSHV), viral gene expression is extremely limited and copy numbers of viral genomes remain constant. Latency-associated nuclear antigen (LANA) is known to have a role in maintaining viral genome copy numbers in growing cells. Several studies have shown that LANA is localized in particular regions on mitotic chromosomes, such as centromeres/pericentromeres. We independently examined the distinct localization of LANA on mitotic chromosomes during mitosis, using super-resolution laser confocal microscopy and correlative fluorescence microscopy-electron microscopy (FM-EM) analyses. We found that the majority of LANA were not localized at particular regions such as telomeres/peritelomeres, centromeres/pericentromeres, and cohesion sites, but at the bodies of condensed chromosomes. Thus, LANA may undergo various interactions with the host factors on the condensed chromosomes in order to tether the viral genome to mitotic chromosomes and realize faithful viral genome segregation during cell division.


Subject(s)
Antigens, Viral/genetics , Chromosome Mapping , Chromosomes, Human , Mitosis/genetics , Nuclear Proteins/genetics , Cell Line , Centromere , Chromosome Mapping/methods , DNA, Viral , Fluorescent Antibody Technique, Indirect , Genome, Viral , Herpesvirus 8, Human/physiology , Humans , Metaphase/genetics , Microscopy, Confocal , Virus Latency
18.
EMBO J ; 32(17): 2336-47, 2013 Aug 28.
Article in English | MEDLINE | ID: mdl-23921551

ABSTRACT

Diverse causes, including pathogenic invasion or the uptake of mineral crystals such as silica and monosodium urate (MSU), threaten cells with lysosomal rupture, which can lead to oxidative stress, inflammation, and apoptosis or necrosis. Here, we demonstrate that lysosomes are selectively sequestered by autophagy, when damaged by MSU, silica, or the lysosomotropic reagent L-Leucyl-L-leucine methyl ester (LLOMe). Autophagic machinery is recruited only on damaged lysosomes, which are then engulfed by autophagosomes. In an autophagy-dependent manner, low pH and degradation capacity of damaged lysosomes are recovered. Under conditions of lysosomal damage, loss of autophagy causes inhibition of lysosomal biogenesis in vitro and deterioration of acute kidney injury in vivo. Thus, we propose that sequestration of damaged lysosomes by autophagy is indispensable for cellular and tissue homeostasis.


Subject(s)
Autophagy/physiology , Kidney Tubules/physiopathology , Lysosomes/metabolism , Animals , Autophagy-Related Protein 7 , Cell Line/drug effects , Dipeptides/pharmacology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Hydrogen-Ion Concentration , Hyperuricemia/physiopathology , Lysosomes/drug effects , Male , Mice , Mice, Transgenic , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , NIH 3T3 Cells/drug effects , Phagosomes/physiology , Uric Acid/pharmacology
19.
Cell Host Microbe ; 12(1): 109-16, 2012 Jul 19.
Article in English | MEDLINE | ID: mdl-22817992

ABSTRACT

Neutrophils contribute to pathogen clearance by producing neutrophil extracellular traps (NETs), which are genomic DNA-based net-like structures that capture bacteria and fungi. Although NETs also express antiviral factors, such as myeloperoxidase and α-defensin, the involvement of NETs in antiviral responses remains unclear. We show that NETs capture human immunodeficiency virus (HIV)-1 and promote HIV-1 elimination through myeloperoxidase and α-defensin. Neutrophils detect HIV-1 by Toll-like receptors (TLRs) TLR7 and TLR8, which recognize viral nucleic acids. Engagement of TLR7 and TLR8 induces the generation of reactive oxygen species that trigger NET formation, leading to NET-dependent HIV-1 elimination. However, HIV-1 counteracts this response by inducing C-type lectin CD209-dependent production of interleukin (IL)-10 by dendritic cells to inhibit NET formation. IL-10 suppresses the reactive oxygen species-dependent generation of NETs induced upon TLR7 and TLR8 engagement, resulting in disrupted NET-dependent HIV-1 elimination. Therefore, NET formation is an antiviral response that is counteracted by HIV-1.


Subject(s)
Extracellular Space/virology , HIV-1/pathogenicity , Host-Pathogen Interactions , Neutrophils/metabolism , Neutrophils/virology , Cell Adhesion Molecules/metabolism , Cells, Cultured , Dendritic Cells/virology , Extracellular Space/metabolism , Humans , Interleukin-10/metabolism , Lectins, C-Type/metabolism , Neutrophils/cytology , Peroxidase/metabolism , Receptors, Cell Surface/metabolism , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/metabolism , alpha-Defensins/metabolism
20.
Cell Host Microbe ; 11(5): 492-503, 2012 May 17.
Article in English | MEDLINE | ID: mdl-22607802

ABSTRACT

Bornaviruses are nonsegmented negative-strand RNA viruses that establish a persistent infection in the nucleus and occasionally integrate a DNA genome copy into the host chromosomal DNA. However, how these viruses achieve intranuclear infection remains unclear. We show that Borna disease virus (BDV), a mammalian bornavirus, closely associates with the cellular chromosome to ensure intranuclear infection. BDV generates viral factories within the nucleus using host chromatin as a scaffold. In addition, the viral ribonucleoprotein (RNP) interacts directly with the host chromosome throughout the cell cycle, using core histones as a docking platform. HMGB1, a host chromatin-remodeling DNA architectural protein, is required to stabilize RNP on chromosomes and for efficient BDV RNA transcription in the nucleus. During metaphase, the association of RNP with mitotic chromosomes allows the viral RNA to segregate into daughter cells and ensure persistent infection. Thus, bornaviruses likely evolved a chromosome-dependent life cycle to achieve stable intranuclear infection.


Subject(s)
Borna disease virus/physiology , Borna disease virus/pathogenicity , Cell Nucleus/virology , Virus Replication , Cell Cycle , Cell Line , Chromosome Segregation , HMGB1 Protein/metabolism , Histones/metabolism , Host-Pathogen Interactions , Humans , Nucleoproteins/metabolism , Protein Binding , RNA, Viral/metabolism , Transcription, Genetic , Viral Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...