Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Cell ; 184(13): 3452-3466.e18, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34139176

ABSTRACT

Antibodies against the receptor-binding domain (RBD) of the SARS-CoV-2 spike protein prevent SARS-CoV-2 infection. However, the effects of antibodies against other spike protein domains are largely unknown. Here, we screened a series of anti-spike monoclonal antibodies from coronavirus disease 2019 (COVID-19) patients and found that some of antibodies against the N-terminal domain (NTD) induced the open conformation of RBD and thus enhanced the binding capacity of the spike protein to ACE2 and infectivity of SARS-CoV-2. Mutational analysis revealed that all of the infectivity-enhancing antibodies recognized a specific site on the NTD. Structural analysis demonstrated that all infectivity-enhancing antibodies bound to NTD in a similar manner. The antibodies against this infectivity-enhancing site were detected at high levels in severe patients. Moreover, we identified antibodies against the infectivity-enhancing site in uninfected donors, albeit at a lower frequency. These findings demonstrate that not only neutralizing antibodies but also enhancing antibodies are produced during SARS-CoV-2 infection.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/immunology , Animals , COVID-19/immunology , Cell Line , Chlorocebus aethiops , HEK293 Cells , Humans , Protein Binding/immunology , Protein Domains/immunology , Spike Glycoprotein, Coronavirus/genetics , Vero Cells
2.
Cell ; 167(2): 382-396.e17, 2016 Oct 06.
Article in English | MEDLINE | ID: mdl-27693356

ABSTRACT

The inflammasome is an intracellular signaling complex, which on recognition of pathogens and physiological aberration, drives activation of caspase-1, pyroptosis, and the release of the pro-inflammatory cytokines IL-1ß and IL-18. Bacterial ligands must secure entry into the cytoplasm to activate inflammasomes; however, the mechanisms by which concealed ligands are liberated in the cytoplasm have remained unclear. Here, we showed that the interferon-inducible protein IRGB10 is essential for activation of the DNA-sensing AIM2 inflammasome by Francisella novicida and contributed to the activation of the LPS-sensing caspase-11 and NLRP3 inflammasome by Gram-negative bacteria. IRGB10 directly targeted cytoplasmic bacteria through a mechanism requiring guanylate-binding proteins. Localization of IRGB10 to the bacterial cell membrane compromised bacterial structural integrity and mediated cytosolic release of ligands for recognition by inflammasome sensors. Overall, our results reveal IRGB10 as part of a conserved signaling hub at the interface between cell-autonomous immunity and innate immune sensing pathways.


Subject(s)
DNA-Binding Proteins/metabolism , Francisella/immunology , GTP Phosphohydrolases/metabolism , Gram-Negative Bacterial Infections/immunology , Host-Pathogen Interactions/immunology , Inflammasomes/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , B-Lymphocytes/immunology , Caspases/metabolism , Caspases, Initiator , Cytosol/immunology , Cytosol/microbiology , GTP Phosphohydrolases/genetics , Gram-Negative Bacterial Infections/microbiology , Immunity, Cellular , Immunity, Innate , Inflammasomes/metabolism , Ligands , Mice , Mice, Mutant Strains , Myeloid Cells/immunology , T-Lymphocytes/immunology
3.
Nat Immunol ; 18(8): 899-910, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28604719

ABSTRACT

Mammalian autophagy-related 8 (Atg8) homologs consist of LC3 proteins and GABARAPs, all of which are known to be involved in canonical autophagy. In contrast, the roles of Atg8 homologs in noncanonical autophagic processes are not fully understood. Here we show a unique role of GABARAPs, in particular gamma-aminobutyric acid (GABA)-A-receptor-associated protein-like 2 (Gabarapl2; also known as Gate-16), in interferon-γ (IFN-γ)-mediated antimicrobial responses. Cells that lacked GABARAPs but not LC3 proteins and mice that lacked Gate-16 alone were defective in the IFN-γ-induced clearance of vacuolar pathogens such as Toxoplasma. Gate-16 but not LC3b specifically associated with the small GTPase ADP-ribosylation factor 1 (Arf1) to mediate uniform distribution of interferon-inducible GTPases. The lack of GABARAPs reduced Arf1 activation, which led to formation of interferon-inducible GTPase-containing aggregates and hampered recruitment of interferon-inducible GTPases to vacuolar pathogens. Thus, GABARAPs are uniquely required for antimicrobial host defense through cytosolic distribution of interferon-inducible GTPases.


Subject(s)
ADP-Ribosylation Factor 1/immunology , Autophagy/immunology , Carrier Proteins/immunology , Interferon-gamma/immunology , Microtubule-Associated Proteins/immunology , Toxoplasma/immunology , Toxoplasmosis/immunology , ADP-Ribosylation Factor 1/metabolism , Animals , Apoptosis Regulatory Proteins , Autophagy-Related Protein 8 Family , CRISPR-Cas Systems , Carrier Proteins/metabolism , Computer Simulation , Cytoskeletal Proteins/immunology , Cytoskeletal Proteins/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , GTP Phosphohydrolases/immunology , GTP Phosphohydrolases/metabolism , Gene Editing , Immunoblotting , Immunoprecipitation , Interferon-gamma/metabolism , Intracellular Signaling Peptides and Proteins , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Microtubule-Associated Proteins/metabolism
4.
Genes Cells ; 29(1): 17-38, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37984375

ABSTRACT

Irgb6 is a priming immune-related GTPase (IRG) that counteracts Toxoplasma gondii. It is known to be recruited to the low virulent type II T. gondii parasitophorous vacuole (PV), initiating cell-autonomous immunity. However, the molecular mechanism by which immunity-related GTPases become inactivated after the parasite infection remains obscure. Here, we found that Thr95 of Irgb6 is prominently phosphorylated in response to low virulent type II T. gondii infection. We observed that a phosphomimetic T95D mutation in Irgb6 impaired its localization to the PV and exhibited reduced GTPase activity in vitro. Structural analysis unveiled an atypical conformation of nucleotide-free Irgb6-T95D, resulting from a conformational change in the G-domain that allosterically modified the PV membrane-binding interface. In silico docking corroborated the disruption of the physiological membrane binding site. These findings provide novel insights into a T. gondii-induced allosteric inactivation mechanism of Irgb6.


Subject(s)
Toxoplasma , Toxoplasma/metabolism , Phosphorylation , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Vacuoles/metabolism
5.
Eur J Immunol ; 53(11): e2350455, 2023 11.
Article in English | MEDLINE | ID: mdl-37471504

ABSTRACT

Caspase activation results in pyroptosis, an inflammatory cell death that contributes to several inflammatory diseases by releasing inflammatory cytokines and cellular contents. Fusobacterium nucleatum is a periodontal pathogen frequently detected in human cancer and inflammatory bowel diseases. Studies have reported that F. nucleatum infection leads to NLRP3 activation and pyroptosis, but the precise activation process and disease association remain poorly understood. This study demonstrated that F. nucleatum infection exacerbates acute colitis in mice and activates pyroptosis through caspase-11-mediated gasdermin D cleavage in macrophages. Furthermore, F. nucleatum infection in colitis mice induces the enhancement of IL-1⍺ secretion from the colon, affecting weight loss and severe disease activities. Neutralization of IL-1⍺ protects F. nucleatum infected mice from severe colitis. Therefore, F. nucleatum infection facilitates inflammation in acute colitis with IL-1⍺ from colon tissue by activating noncanonical inflammasome through gasdermin D cleavage.


Subject(s)
Colitis , Inflammasomes , Humans , Animals , Mice , Inflammasomes/metabolism , Fusobacterium nucleatum/metabolism , Gasdermins , Colitis/chemically induced , Caspases/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
6.
J Infect Chemother ; 28(4): 486-491, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34930624

ABSTRACT

INTRODUCTION: The aims were to investigate the clinical characteristics of Toxoplasma gondii (T. gondii) immunoglobulin (Ig) M-positive mothers and to clarify the incidences of serum T. gondii IgM or blood T. gondii DNA positivity in newborns born to the mothers and the actual congenital T. gondii infection. METHODS: Mothers with T. gondii IgM positivity and newborns born to the mothers from 2013 to 2020 were prospectively investigated. Serum T. gondii IgG and IgM were measured by enzyme-linked immunosorbent assay. Blood T. gondii DNA was detected by semi-nested polymerase chain reaction. Congenital T. gondii infection was diagnosed based on clinical characteristic manifestations with serum T. gondii IgG positivity at any age or T. gondii IgG positivity after 12 months of age. RESULTS: Among 71 T. gondii IgM-positive mothers, including one with triplets, 41% had low T. gondii IgG avidity index and 73% received maternal therapy. Among 73 newborns who were examined for serum T. gondii IgG and IgM at birth, none had clinical manifestations, and one (1.4%) had T. gondii IgM positivity. Among 32 newborns who were examined for blood T. gondii DNA at birth, two (6.3%) were positive. All patients with serum T. gondii IgM or blood T. gondii DNA positivity showed T. gondii IgG negativity within 12 months of age. CONCLUSIONS: A few newborns born to T. gondii IgM-positive mothers were suspected of having congenital T. gondii infection based on serum T. gondii IgM or blood T. gondii DNA testing at birth. However, none developed congenital T. gondii infection.


Subject(s)
Toxoplasma , Antibodies, Protozoan , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunoglobulin M , Infant, Newborn , Mothers , Pregnancy , Prospective Studies , Toxoplasma/genetics
7.
Immunity ; 36(5): 782-94, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22560444

ABSTRACT

Effective major histocompatibility complex-II (MHC-II) antigen presentation from phagocytosed particles requires phagosome-intrinsic Toll-like receptor (TLR) signaling, but the molecular mechanisms underlying TLR delivery to phagosomes and how signaling regulates antigen presentation are incompletely understood. We show a requirement in dendritic cells (DCs) for adaptor protein-3 (AP-3) in efficient TLR recruitment to phagosomes and MHC-II presentation of antigens internalized by phagocytosis but not receptor-mediated endocytosis. DCs from AP-3-deficient pearl mice elicited impaired CD4(+) T cell activation and Th1 effector cell function to particulate antigen in vitro and to recombinant Listeria monocytogenes infection in vivo. Whereas phagolysosome maturation and peptide:MHC-II complex assembly proceeded normally in pearl DCs, peptide:MHC-II export to the cell surface was impeded. This correlated with reduced TLR4 recruitment and proinflammatory signaling from phagosomes by particulate TLR ligands. We propose that AP-3-dependent TLR delivery from endosomes to phagosomes and subsequent signaling mobilize peptide:MHC-II export from intracellular stores.


Subject(s)
Adaptor Protein Complex 3/immunology , Antigen Presentation/immunology , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Phagosomes/immunology , Toll-Like Receptors/immunology , Adaptor Protein Complex 3/metabolism , Animals , Antigens/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Cell Membrane/immunology , Cell Membrane/metabolism , Cells, Cultured , Dendritic Cells/metabolism , Endocytosis/immunology , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Ligands , Listeria monocytogenes/immunology , Listeriosis/immunology , Listeriosis/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/metabolism , Ovalbumin/immunology , Ovalbumin/metabolism , Peptides/immunology , Peptides/metabolism , Phagocytosis/immunology , Phagosomes/metabolism , Signal Transduction/immunology , Th1 Cells/immunology , Th1 Cells/metabolism , Toll-Like Receptors/metabolism
8.
Immunity ; 37(6): 986-997, 2012 Dec 14.
Article in English | MEDLINE | ID: mdl-23219390

ABSTRACT

Toll-like receptor-9 (TLR9) is largely responsible for discriminating self from pathogenic DNA. However, association of host DNA with autoantibodies activates TLR9, inducing the pathogenic secretion of type I interferons (IFNs) from plasmacytoid dendritic cells (pDCs). Here, we found that in response to DNA-containing immune complexes (DNA-IC), but not to soluble ligands, IFN-α production depended upon the convergence of the phagocytic and autophagic pathways, a process called microtubule-associated protein 1A/1B-light chain 3 (LC3)-associated phagocytosis (LAP). LAP was required for TLR9 trafficking into a specialized interferon signaling compartment by a mechanism that involved autophagy-related proteins, but not the conventional autophagic preinitiation complex, or adaptor protein-3 (AP-3). Our findings unveil a new role for nonconventional autophagy in inflammation and provide one mechanism by which anti-DNA autoantibodies, such as those found in several autoimmune disorders, bypass the controls that normally restrict the apportionment of pathogenic DNA and TLR9 to the interferon signaling compartment.


Subject(s)
Antigen-Antibody Complex/immunology , Autophagy/immunology , DNA/immunology , Interferon Type I/biosynthesis , Animals , Humans , Immunoglobulin G/immunology , Membrane Transport Proteins/metabolism , Mice , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Phagocytosis/immunology , Phagosomes/metabolism , Protein Transport , Toll-Like Receptor 9/immunology , Toll-Like Receptor 9/metabolism
9.
Immunity ; 37(2): 302-13, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22795875

ABSTRACT

Interferon-γ (IFN-γ) is essential for host defense against intracellular pathogens. Stimulation of innate immune cells by IFN-γ upregulates ∼2,000 effector genes such as immunity-related GTPases including p65 guanylate-binding protein (Gbp) family genes. We show that a cluster of Gbp genes was required for host cellular immunity against the intracellular parasite Toxoplasma gondii. We generated mice deficient for all six Gbp genes located on chromosome 3 (Gbp(chr3)) by targeted chromosome engineering. Mice lacking Gbp(chr3) were highly susceptible to T. gondii infection, resulting in increased parasite burden in immune organs. Furthermore, Gbp(chr3)-deleted macrophages were defective in IFN-γ-mediated suppression of T. gondii intracellular growth and recruitment of IFN-γ-inducible p47 GTPase Irgb6 to the parasitophorous vacuole. In addition, some members of Gbp(chr3) restored the protective response against T. gondii in Gbp(chr3)-deleted cells. Our results suggest that Gbp(chr3) play a pivotal role in anti-T. gondii host defense by controlling IFN-γ-mediated Irgb6-dependent cellular innate immunity.


Subject(s)
GTP-Binding Proteins/immunology , Immunity, Innate/immunology , Interferon-gamma/immunology , Macrophages/immunology , Toxoplasma/immunology , Toxoplasmosis/immunology , Animals , Chromosomes, Mammalian/genetics , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Humans , Immunity, Cellular/immunology , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/metabolism , Listeria monocytogenes/immunology , Luminescent Measurements , Macrophages/metabolism , Macrophages/ultrastructure , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Nitric Oxide/metabolism , Toxoplasma/metabolism
10.
Immunity ; 35(1): 3-5, 2011 Jul 22.
Article in English | MEDLINE | ID: mdl-21777792

ABSTRACT

Unc93B1, a multitransmembrane ER-resident protein, controls intracellular trafficking of endosomal Toll-like receptors. In this issue of Immunity, Fukui et al. (2011) revealed that Unc93B1 regulates differential transport of TLR7 and TLR9 into signaling endosomes to prevent autoimmunity.

11.
Proc Natl Acad Sci U S A ; 114(50): E10782-E10791, 2017 12 12.
Article in English | MEDLINE | ID: mdl-29187532

ABSTRACT

Signal peptide peptidase (SPP) is an intramembrane aspartic protease involved in the maturation of the core protein of hepatitis C virus (HCV). The processing of HCV core protein by SPP has been reported to be critical for the propagation and pathogenesis of HCV. Here we examined the inhibitory activity of inhibitors for γ-secretase, another intramembrane cleaving protease, against SPP, and our findings revealed that the dibenzoazepine-type structure in the γ-secretase inhibitors is critical for the inhibition of SPP. The spatial distribution showed that the γ-secretase inhibitor compound YO-01027 with the dibenzoazepine structure exhibits potent inhibiting activity against SPP in vitro and in vivo through the interaction of Val223 in SPP. Treatment with this SPP inhibitor suppressed the maturation of core proteins of all HCV genotypes without the emergence of drug-resistant viruses, in contrast to the treatment with direct-acting antivirals. YO-01027 also efficiently inhibited the propagation of protozoa such as Plasmodium falciparum and Toxoplasma gondii These data suggest that SPP is an ideal target for the development of therapeutics not only against chronic hepatitis C but also against protozoiasis.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Antiprotozoal Agents/pharmacology , Antiviral Agents/pharmacology , Aspartic Acid Endopeptidases/antagonists & inhibitors , Dibenzazepines/pharmacology , Hepacivirus/drug effects , Protease Inhibitors/pharmacology , Animals , Antiprotozoal Agents/chemistry , Antiviral Agents/chemistry , Cell Line , Dibenzazepines/chemistry , HEK293 Cells , Hepacivirus/genetics , Humans , Mice , Mice, Inbred BALB C , Models, Molecular , Plasmodium falciparum/drug effects , Protease Inhibitors/chemistry , Structure-Activity Relationship , Toxoplasma/drug effects , Viral Core Proteins/antagonists & inhibitors , Virus Replication/drug effects
12.
Int Immunol ; 30(3): 113-119, 2018 03 10.
Article in English | MEDLINE | ID: mdl-29408976

ABSTRACT

Toxoplasma gondii can infect homoeothermic animals including humans and cause lethal toxoplasmosis in immunocompromised individuals. When hosts are infected with T. gondii, the cells induce immune responses against T. gondii. The pathogen infection is recognized by immune sensors that directly detect T. gondii structural components, leading to production of pro-inflammatory cytokines and chemokines. Antigen-presenting cells such as macrophages and dendritic cells strongly activate T cells and induce development of Th1 cells and antigen-specific killer CD8 T cells. These T cells and Group 1 innate lymphoid cells are main producers of IFN-γ, which robustly stimulates cell-autonomous immunity in cells infected with T. gondii. IFN-γ-inducible effectors such as IFN-inducible GTPases, inducible nitric oxide synthase and indoleamine-2,3-dioxygenase differentially play important roles in suppression of T. gondii growth and its direct killing in anti-T. gondii cell-autonomous immune responses. In this review, we will describe our current knowledge of innate, adaptive and IFN-γ-mediated cell-autonomous immunity against T. gondii infection.


Subject(s)
Immunity, Cellular/immunology , Interferon-gamma/immunology , Toxoplasma/immunology , Toxoplasmosis/immunology , Toxoplasmosis/parasitology , Animals , Host-Pathogen Interactions/immunology , Humans
13.
Proc Natl Acad Sci U S A ; 112(33): E4581-90, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26240314

ABSTRACT

IFN-γ orchestrates cell-autonomous host defense against various intracellular vacuolar pathogens. IFN-γ-inducible GTPases, such as p47 immunity-related GTPases (IRGs) and p65 guanylate-binding proteins (GBPs), are recruited to pathogen-containing vacuoles, which is important for disruption of the vacuoles, culminating in the cell-autonomous clearance. Although the positive regulation for the proper recruitment of IRGs and GBPs to the vacuoles has been elucidated, the suppressive mechanism is unclear. Here, we show that Rab GDP dissociation inhibitor α (RabGDIα), originally identified as a Rab small GTPase inhibitor, is a negative regulator of IFN-γ-inducible GTPases in cell-autonomous immunity to the intracellular pathogen Toxoplasma gondii. Overexpression of RabGDIα, but not of RabGDIß, impaired IFN-γ-dependent reduction of T. gondii numbers. Conversely, RabGDIα deletion in macrophages and fibroblasts enhanced the IFN-γ-induced clearance of T. gondii. Furthermore, upon a high dose of infection by T. gondii, RabGDIα-deficient mice exhibited a decreased parasite burden in the brain and increased resistance in the chronic phase than did control mice. Among members of IRGs and GBPs important for the parasite clearance, Irga6 and Gbp2 alone were more frequently recruited to T. gondii-forming parasitophorous vacuoles in RabGDIα-deficient cells. Notably, Gbp2 positively controlled Irga6 recruitment that was inhibited by direct and specific interactions of RabGDIα with Gbp2 through the lipid-binding pocket. Taken together, our results suggest that RabGDIα inhibits host defense against T. gondii by negatively regulating the Gbp2-Irga6 axis of IFN-γ-dependent cell-autonomous immunity.


Subject(s)
GTP Phosphohydrolases/metabolism , Gene Expression Regulation, Enzymologic , Guanine Nucleotide Dissociation Inhibitors/metabolism , Interferon-gamma/immunology , Toxoplasma/pathogenicity , Toxoplasmosis/immunology , Amino Acid Sequence , Animals , Base Sequence , Chlorocebus aethiops , DNA Primers/genetics , Female , Fibroblasts/metabolism , Inflammation/immunology , Lipids/chemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Protein Binding , Sequence Homology, Amino Acid , Vero Cells
14.
Int Immunol ; 27(9): 435-45, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25925170

ABSTRACT

Toll-like receptors (TLRs) traffic to distinct membranes for signaling. TLR7 and TLR9 recognize viral nucleic acids in the endosomes and induce robust anti-viral program. Signaling from these TLRs bifurcate at the level of distinct endosomal compartments, namely VAMP3(+) and LAMP(+) endosomes, to mediate the induction of cytokine and type I interferon (IFN) genes, respectively. The formation of the TLR9 endosome competent for IFNs induction requires AP-3. Phosphoinositides (PIs) mark distinct subcellular membranes and control membrane trafficking. However, their role in TLR trafficking and signaling in different dendritic cell (DC) subsets remains unclear. Here, we examined the role of phosphatidylinositol 3P 5-kinase, PIKfyve, in TLR9 trafficking and signaling. We demonstrate that inhibition of PIKfyve activity preferentially blocks TLR9 signaling for type I IFN induction in FLT3L-bone marrow-derived DCs. By confocal microscopy using RAW264.7 cells, we show that trafficking of both TLR9 and CpG to the LAMP1(+) compartment was blocked by PIKfyve inhibitor treatment, whereas their trafficking to the VAMP3(+) endosome remained intact. Further, AP-3 recruitment to TLR9 endosomes was impaired by PIKfyve inhibition. These data indicate that PIKfyve provides critical PIs necessary for the formation of endosome from which TLR9 signals to induce type I IFNs.


Subject(s)
Interferon Type I/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/physiology , Toll-Like Receptor 9/metabolism , Animals , Cell Line , Cell Membrane/metabolism , DNA-Binding Proteins/metabolism , Dendritic Cells/metabolism , Endosomes/metabolism , Lysosomal Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Protein Transport/physiology , Toll-Like Receptor 7/metabolism , Transcription Factors/metabolism , Vesicle-Associated Membrane Protein 3/metabolism
15.
J Immunol ; 192(7): 3328-35, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24563254

ABSTRACT

IFN-γ mediates cellular innate immunity against an intracellular parasite, Toxoplasma gondii, by inducing immunity-related GTPases such as p47 IFN-γ-regulated GTPases (IRGs) and p65 guanylate-binding proteins (GBPs), which also participate in antibacterial responses via autophagy. An essential autophagy protein, Atg5, was previously shown to play a critical role in anti-T. gondii cell-autonomous immunity. However, the involvement of other autophagy proteins remains unknown. In this study, we show that essential autophagy proteins differentially participate in anti-T. gondii cellular immunity by recruiting IFN-γ-inducible GTPases. IFN-γ-induced suppression of T. gondii proliferation and recruitment of an IRG Irgb6 and GBPs are profoundly impaired in Atg7- or Atg16L1-deficient cells. In contrast, cells lacking other essential autophagy proteins, Atg9a and Atg14, are capable of mediating the anti-T. gondii response and recruiting Irgb6 and GBPs to the parasites. Although IFN-γ also stimulates anti-T. gondii cellular immunity in humans, whether this response requires GBPs and human autophagy proteins remains to be seen. To analyze the role of human ATG16L1 and GBPs in IFN-γ-mediated anti-T. gondii responses, human cells lacking ATG16L1 or GBPs were generated by the Cas9/CRISPR genome-editing technique. Although both ATG16L1 and GBPs are dispensable for IFN-γ-induced inhibition of T. gondii proliferation in the human cells, human ATG16L1 is also required for the recruitment of GBPs. Taken together, human ATG16L1 and mouse autophagy components Atg7 and Atg16L1, but not Atg9a and Atg14, participate in the IFN-γ-induced recruitment of the immunity-related GTPases to the intracellular pathogen.


Subject(s)
Autophagy/immunology , Carrier Proteins/immunology , Interferon-gamma/immunology , Microtubule-Associated Proteins/immunology , Toxoplasma/immunology , Animals , Autophagy/genetics , Autophagy-Related Protein 7 , Autophagy-Related Proteins , Base Sequence , Blotting, Western , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Cells, Cultured , Chlorocebus aethiops , Embryo, Mammalian/cytology , Fibroblasts/immunology , Fibroblasts/metabolism , Fibroblasts/parasitology , Gene Knockout Techniques , Host-Parasite Interactions/immunology , Humans , Immunity, Cellular/genetics , Immunity, Cellular/immunology , Immunity, Innate/genetics , Immunity, Innate/immunology , Interferon-gamma/metabolism , Mice , Mice, Knockout , Microscopy, Confocal , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Molecular Sequence Data , Toxoplasma/physiology , Vero Cells
16.
J Virol ; 88(4): 2157-67, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24335288

ABSTRACT

The baculovirus Autographa californica nucleopolyhedrovirus (AcNPV) has been widely used to achieve a high level of foreign gene expression in insect cells, as well as for efficient gene transduction into mammalian cells without any replication. In addition to permitting efficient gene delivery, baculovirus has been shown to induce host innate immune responses in various mammalian cells and in mice. In this study, we examined the effects of the innate immune responses on gene expression by recombinant baculoviruses in cultured cells. The reporter gene expression in IRF3-deficient mouse embryonic fibroblasts (MEFs) infected with the recombinant baculovirus was shown to be enhanced in accordance with the suppression of beta interferon (IFN-ß) production. Furthermore, efficient gene transduction by the recombinant baculovirus was achieved in MEFs deficient for stimulator of interferon genes (STING), TANK binding kinase 1 (TBK1), IFN regulatory factor 3 (IRF3), or IFN-ß promoter stimulator 1 (IPS-1), but not in those deficient for IRF7, MyD88, or Z-DNA binding protein 1 (ZBP1)/DAI. Enhancement of gene expression by the recombinant baculovirus was also observed in human hepatoma cell lines replicating hepatitis C virus (HCV), in which innate immunity was impaired by the cleavage of IPS-1 by the viral protease. In addition, infection with the recombinant baculovirus expressing the BH3-only protein, BIMS, a potent inducer of apoptosis, resulted in a selective cell death in the HCV replicon cells. These results indicate that innate immune responses induced by infection with baculovirus attenuate transgene expression, and this characteristic might be useful for a selective gene transduction into cells with impaired innate immunity arising from infection with various viruses.


Subject(s)
Gene Expression Regulation/immunology , Immunity, Innate/immunology , Nucleopolyhedroviruses/immunology , Transgenes/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line , DNA Primers/genetics , Fibroblasts/metabolism , Fluorescent Antibody Technique , Humans , Immunoblotting , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Moths/virology , Real-Time Polymerase Chain Reaction , Transduction, Genetic
17.
Cell Rep ; 43(5): 114131, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38656870

ABSTRACT

Atg8 paralogs, consisting of LC3A/B/C and GBRP/GBRPL1/GATE16, function in canonical autophagy; however, their function is controversial because of functional redundancy. In innate immunity, xenophagy and non-canonical single membranous autophagy called "conjugation of Atg8s to single membranes" (CASM) eliminate bacteria in various cells. Previously, we reported that intracellular Streptococcus pneumoniae can induce unique hierarchical autophagy comprised of CASM induction, shedding, and subsequent xenophagy. However, the molecular mechanisms underlying these processes and the biological significance of transient CASM induction remain unknown. Herein, we profile the relationship between Atg8s, autophagy receptors, poly-ubiquitin, and Atg4 paralogs during pneumococcal infection to understand the driving principles of hierarchical autophagy and find that GATE16 and GBRP sequentially play a pivotal role in CASM shedding and subsequent xenophagy induction, respectively, and LC3A and GBRPL1 are involved in CASM/xenophagy induction. Moreover, we reveal ingenious bacterial tactics to gain intracellular survival niches by manipulating CASM-xenophagy progression by generating intracellular pneumococci-derived H2O2.


Subject(s)
Autophagy-Related Protein 8 Family , Streptococcus pneumoniae , Animals , Mice , Autophagy , Autophagy-Related Protein 8 Family/metabolism , Autophagy-Related Proteins/metabolism , Macroautophagy , Microtubule-Associated Proteins/metabolism , Pneumococcal Infections/microbiology , Pneumococcal Infections/metabolism , Pneumococcal Infections/immunology , Streptococcus pneumoniae/metabolism
18.
Nat Commun ; 15(1): 4278, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38778039

ABSTRACT

Toxoplasma gondii is a global protozoan pathogen. Clonal lineages predominate in Europe, North America, Africa, and China, whereas highly recombinant parasites are endemic in South/Central America. Far East Asian T. gondii isolates are not included in current global population genetic structure analyses at WGS resolution. Here we report a genome-wide population study that compared eight Japanese and two Chinese isolates against representative worldwide T. gondii genomes using POPSICLE, a novel population structure analyzing software. Also included were 7 genomes resurrected from non-viable isolates by target enrichment sequencing. Visualization of the genome structure by POPSICLE shows a mixture of Chinese haplogroup (HG) 13 haploblocks introgressed within the genomes of Japanese HG2 and North American HG12. Furthermore, two ancestral lineages were identified in the Japanese strains; one lineage shares a common ancestor with HG11 found in both Japanese strains and North American HG12. The other ancestral lineage, found in T. gondii isolates from a small island in Japan, is admixed with genetically diversified South/Central American strains. Taken together, this study suggests multiple ancestral links between Far East Asian and American T. gondii strains and provides insight into the transmission history of this cosmopolitan organism.


Subject(s)
Genome, Protozoan , Phylogeny , Toxoplasma , Toxoplasma/genetics , Toxoplasma/classification , Humans , North America , Genome, Protozoan/genetics , Toxoplasmosis/parasitology , China , Central America , Japan , Haplotypes , Genetic Variation , Recombination, Genetic
19.
Cell Rep ; 42(6): 112592, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37269286

ABSTRACT

Secreted virulence factors of Toxoplasma to survive in immune-competent hosts have been extensively explored by classical genetics and in vivo CRISPR screen methods, whereas their requirements in immune-deficient hosts are incompletely understood. Those of non-secreted virulence factors are further enigmatic. Here we develop an in vivo CRISPR screen system to enrich not only secreted but also non-secreted virulence factors in virulent Toxoplasma-infected C57BL/6 mice. Notably, combined usage of immune-deficient Ifngr1-/- mice highlights genes encoding various non-secreted proteins as well as well-known effectors such as ROP5, ROP18, GRA12, and GRA45 as interferon-γ (IFN-γ)-dependent virulence genes. The screen results suggest a role of GRA72 for normal GRA17/GRA23 localization and the IFN-γ-dependent role of UFMylation-related genes. Collectively, our study demonstrates that host genetics can complement in vivo CRISPR screens to highlight genes encoding IFN-γ-dependent secreted and non-secreted virulence factors in Toxoplasma.


Subject(s)
Toxoplasma , Virulence Factors , Animals , Mice , Virulence Factors/genetics , Virulence Factors/metabolism , Toxoplasma/metabolism , Interferon-gamma/genetics , Protozoan Proteins/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Mice, Inbred C57BL
20.
mBio ; 14(1): e0325622, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36715543

ABSTRACT

Toxoplasma gondii secretes various virulence effector molecules into host cells to disrupt host interferon-γ (IFN-γ)-dependent immunity. Among these effectors, ROP18 directly phosphorylates and inactivates IFN-inducible GTPases, such as immunity-related GTPases (IRGs) and guanylate-binding proteins (GBPs), leading to the subversion of IFN-inducible GTPase-induced cell-autonomous immunity. The modes of action of ROP18 have been studied extensively; however, little is known about the molecular mechanisms by which ROP18 is produced in the parasite itself. Here, we report the role of T. gondii transcription factor IWS1 in ROP18 mRNA expression in the parasite. Compared with wild-type virulent type I T. gondii, IWS1-deficient parasites showed dramatically increased loading of IRGs and GBPs onto the parasitophorous vacuole membrane (PVM). Moreover, IWS1-deficient parasites displayed decreased virulence in wild-type mice but retained normal virulence in mice lacking the IFN-γ receptor. Furthermore, IWS1-deficient parasites showed severely decreased ROP18 mRNA expression; however, tagged IWS1 did not directly bind with genomic regions of the ROP18 locus. Ectopic expression of ROP18 in IWS1-deficient parasites restored the decreased loading of effectors onto the PVM and in vivo virulence in wild-type mice. Taken together, these data demonstrate that T. gondii IWS1 indirectly regulates ROP18 mRNA expression to determine fitness in IFN-γ-activated host cells and mice. IMPORTANCE The parasite Toxoplasma gondii has a counterdefense system against interferon-γ (IFN-γ)-dependent host immunity which relies on the secretion of parasite effector proteins. ROP18 is one of the effector, which is released into host cells to inactivate IFN-γ-dependent anti-Toxoplasma host proteins. The mechanism by which Toxoplasma ROP18 subverts host immunity has been extensively analyzed, but how Toxoplasma produces this virulence factor remains unclear. Here, we show that Toxoplasma transcription factor IWS1 is important for ROP18 mRNA expression in the parasite. Loss of IWS1 from virulent Toxoplasma leads to dramatically decreased ROP18 mRNA expression, resulting in profoundly decreased virulence due to greater activity of IFN-γ-dependent host immune responses. Thus, Toxoplasma prepares the critical virulence factor ROP18 via an IWS1-dependent system to negate IFN-γ-dependent antiparasitic immunity and thus survive in the host.


Subject(s)
Toxoplasma , Animals , Mice , Carrier Proteins , GTP Phosphohydrolases/metabolism , Interferon-gamma , Protozoan Proteins/metabolism , Virulence Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL