Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Nat Immunol ; 17(11): 1252-1262, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27595231

ABSTRACT

The mammalian cytoplasmic multi-tRNA synthetase complex (MSC) is a depot system that regulates non-translational cellular functions. Here we found that the MSC component glutamyl-prolyl-tRNA synthetase (EPRS) switched its function following viral infection and exhibited potent antiviral activity. Infection-specific phosphorylation of EPRS at Ser990 induced its dissociation from the MSC, after which it was guided to the antiviral signaling pathway, where it interacted with PCBP2, a negative regulator of mitochondrial antiviral signaling protein (MAVS) that is critical for antiviral immunity. This interaction blocked PCBP2-mediated ubiquitination of MAVS and ultimately suppressed viral replication. EPRS-haploid (Eprs+/-) mice showed enhanced viremia and inflammation and delayed viral clearance. This stimulus-inducible activation of MAVS by EPRS suggests an unexpected role for the MSC as a regulator of immune responses to viral infection.


Subject(s)
Amino Acyl-tRNA Synthetases/metabolism , Disease Resistance/immunology , Host-Pathogen Interactions/immunology , Virus Diseases/immunology , Virus Diseases/metabolism , Amino Acyl-tRNA Synthetases/chemistry , Amino Acyl-tRNA Synthetases/genetics , Animals , Antiviral Agents/pharmacology , Disease Models, Animal , Immunity, Innate , Mice , Mice, Knockout , Peptides/pharmacology , Phosphorylation , Protein Binding , RNA Virus Infections/immunology , RNA Virus Infections/metabolism , RNA Virus Infections/virology , RNA Viruses/drug effects , RNA Viruses/immunology , RNA-Binding Proteins/metabolism , Signal Transduction , Ubiquitination , Virus Diseases/virology , Virus Replication
2.
J Biol Chem ; 293(47): 18110-18122, 2018 11 23.
Article in English | MEDLINE | ID: mdl-30282804

ABSTRACT

Multifunctional autoprocessing repeats-in-toxin (MARTX) toxins are secreted by Gram-negative bacteria and function as primary virulence-promoting macromolecules that deliver multiple cytopathic and cytotoxic effector domains into the host cytoplasm. Among these effectors, Ras/Rap1-specific endopeptidase (RRSP) catalyzes the sequence-specific cleavage of the Switch I region of the cellular substrates Ras and Rap1 that are crucial for host innate immune defenses during infection. To dissect the molecular basis underpinning RRSP-mediated substrate inactivation, we determined the crystal structure of an RRSP from the sepsis-causing bacterial pathogen Vibrio vulnificus (VvRRSP). Structural and biochemical analyses revealed that VvRRSP is a metal-independent TIKI family endopeptidase composed of an N-terminal membrane-localization and substrate-recruitment domain (N lobe) connected via an inter-lobe linker to the C-terminal active site-coordinating core ß-sheet-containing domain (C lobe). Structure-based mutagenesis identified the 2His/2Glu catalytic residues in the core catalytic domain that are shared with other TIKI family enzymes and that are essential for Ras processing. In vitro KRas cleavage assays disclosed that deleting the N lobe in VvRRSP causes complete loss of enzymatic activity. Endogenous Ras cleavage assays combined with confocal microscopy analysis of HEK293T cells indicated that the N lobe functions both in membrane localization via the first α-helix and in substrate assimilation by altering the functional conformation of the C lobe to facilitate recruitment of cellular substrates. Collectively, these results indicate that RRSP is a critical virulence factor that robustly inactivates Ras and Rap1 and augments the pathogenicity of invading bacteria via the combined effects of its N and C lobes.


Subject(s)
Bacterial Proteins/metabolism , Endopeptidases/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Sepsis/enzymology , Sepsis/microbiology , Vibrio vulnificus/enzymology , rap1 GTP-Binding Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Toxins , Endopeptidases/chemistry , Endopeptidases/genetics , HEK293 Cells , Host-Pathogen Interactions , Humans , Protein Domains , Proto-Oncogene Proteins p21(ras)/chemistry , Proto-Oncogene Proteins p21(ras)/genetics , Sepsis/genetics , Vibrio vulnificus/chemistry , Vibrio vulnificus/genetics , rap1 GTP-Binding Proteins/chemistry , rap1 GTP-Binding Proteins/genetics
3.
PLoS Pathog ; 11(9): e1005192, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26406498

ABSTRACT

A transcriptome analysis identified Vibrio vulnificus cabABC genes which were preferentially expressed in biofilms. The cabABC genes were transcribed as a single operon. The cabA gene was induced by elevated 3',5'-cyclic diguanylic acid (c-di-GMP) and encoded a calcium-binding protein CabA. Comparison of the biofilms produced by the cabA mutant and its parent strain JN111 in microtiter plates using crystal-violet staining demonstrated that CabA contributed to biofilm formation in a calcium-dependent manner under elevated c-di-GMP conditions. Genetic and biochemical analyses revealed that CabA was secreted to the cell exterior through functional CabB and CabC, distributed throughout the biofilm matrix, and produced as the biofilm matured. These results, together with the observation that CabA also contributes to the development of rugose colony morphology, indicated that CabA is a matrix-associated protein required for maturation, rather than adhesion involved in the initial attachment, of biofilms. Microscopic comparison of the structure of biofilms produced by JN111 and the cabA mutant demonstrated that CabA is an extracellular matrix component essential for the development of the mature biofilm structures in flow cells and on oyster shells. Exogenously providing purified CabA restored the biofilm- and rugose colony-forming abilities of the cabA mutant when calcium was available. Circular dichroism and size exclusion analyses revealed that calcium binding induces CabA conformational changes which may lead to multimerization. Extracellular complementation experiments revealed that CabA can assemble a functional matrix only when exopolysaccharides coexist. Consequently, the combined results suggested that CabA is a structural protein of the extracellular matrix and multimerizes to a conformation functional in building robust biofilms, which may render V. vulnificus to survive in hostile environments and reach a concentrated infective dose.


Subject(s)
Biofilms , Genes, Bacterial , Polysaccharides, Bacterial/genetics , Vibrio vulnificus/physiology , Amino Acid Sequence , Calcium-Binding Proteins/metabolism , Humans , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Operon , Polysaccharides, Bacterial/metabolism , Real-Time Polymerase Chain Reaction
4.
PLoS Pathog ; 11(10): e1005252, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26496079

ABSTRACT

[This corrects the article DOI: 10.1371/journal.ppat.1005192.].

5.
Proc Natl Acad Sci U S A ; 110(30): E2829-37, 2013 Jul 23.
Article in English | MEDLINE | ID: mdl-23832782

ABSTRACT

Pathogenic and commensal bacteria that experience limited nutrient availability in their host have evolved sophisticated systems to catabolize the mucin sugar N-acetylneuraminic acid, thereby facilitating their survival and colonization. The correct function of the associated catabolic machinery is particularly crucial for the pathogenesis of enteropathogenic bacteria during infection, although the molecular mechanisms involved with the regulation of the catabolic machinery are unknown. This study reports the complex structure of NanR, a repressor of the N-acetylneuraminate (nan) genes responsible for N-acetylneuraminic acid catabolism, and its regulatory ligand, N-acetylmannosamine 6-phosphate (ManNAc-6P), in the human pathogenic bacterium Vibrio vulnificus. Structural studies combined with electron microscopic, biochemical, and in vivo analysis demonstrated that NanR forms a dimer in which the two monomers create an arched tunnel-like DNA-binding space, which contains positively charged residues that interact with the nan promoter. The interaction between the NanR dimer and DNA is alleviated by the ManNAc-6P-mediated relocation of residues in the ligand-binding domain of NanR, which subsequently relieves the repressive effect of NanR and induces the transcription of the nan genes. Survival studies in which mice were challenged with a ManNAc-6P-binding-defective mutant strain of V. vulnificus demonstrated that this relocation of NanR residues is critical for V. vulnificus pathogenesis. In summary, this study presents a model of the mechanism that regulates sialic acid catabolism via NanR in V. vulnificus.


Subject(s)
N-Acetylneuraminic Acid/metabolism , Repressor Proteins/metabolism , Transcription, Genetic , Vibrio vulnificus/metabolism , Microscopy, Electron , Models, Molecular
6.
Nat Commun ; 15(1): 6218, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39043696

ABSTRACT

Multiple bacterial genera take advantage of the multifunctional autoprocessing repeats-in-toxin (MARTX) toxin to invade host cells. Secretion of the MARTX toxin by Vibrio vulnificus, a deadly opportunistic pathogen that causes primary septicemia, the precursor of sepsis, is a major driver of infection; however, the molecular mechanism via which the toxin contributes to septicemia remains unclear. Here, we report the crystal and cryo-electron microscopy (EM) structures of a toxin effector duet comprising the domain of unknown function in the first position (DUF1)/Rho inactivation domain (RID) complexed with human targets. These structures reveal how the duet is used by bacteria as a potent weapon. The data show that DUF1 acts as a RID-dependent transforming NADase domain (RDTND) that disrupts NAD+ homeostasis by hijacking calmodulin. The cryo-EM structure of the RDTND-RID duet complexed with calmodulin and Rac1, together with immunological analyses in vitro and in mice, provide mechanistic insight into how V. vulnificus uses the duet to suppress ROS generation by depleting NAD(P)+ and modifying Rac1 in a mutually-reinforcing manner that ultimately paralyzes first line immune responses, promotes dissemination of invaders, and induces sepsis. These data may allow development of tools or strategies to combat MARTX toxin-related human diseases.


Subject(s)
Bacterial Toxins , Cryoelectron Microscopy , Vibrio vulnificus , Vibrio vulnificus/metabolism , Vibrio vulnificus/pathogenicity , Animals , Humans , Mice , Bacterial Toxins/metabolism , Bacterial Toxins/chemistry , Female , NAD/metabolism , Reactive Oxygen Species/metabolism , Sepsis/microbiology , Protein Domains , Vibrio Infections/microbiology , NAD+ Nucleosidase/metabolism , NAD+ Nucleosidase/chemistry , Crystallography, X-Ray
7.
Mol Cell Biol ; 43(5): 223-240, 2023.
Article in English | MEDLINE | ID: mdl-37154023

ABSTRACT

Glutamyl-prolyl-tRNA synthetase 1 (EPRS1) is known to associated with fibrosis through its catalytic activity to produce prolyl-tRNA. Although its catalytic inhibitor halofuginone (HF) has been known to inhibit the TGF-ß pathway as well as to reduce prolyl-tRNA production for the control of fibrosis, the underlying mechanism how EPRS1 regulates the TGF-ß pathway was not fully understood. Here, we show a noncatalytic function of EPRS1 in controlling the TGF-ß pathway and hepatic stellate cell activation via its interaction with TGF-ß receptor I (TßRI). Upon stimulation with TGF-ß, EPRS1 is phosphorylated by TGF-ß-activated kinase 1 (TAK1), leading to its dissociation from the multi-tRNA synthetase complex and subsequent binding with TßRI. This interaction increases the association of TßRI with SMAD2/3 while decreases that of TßRI with SMAD7. Accordingly, EPRS1 stabilizes TßRI by preventing the ubiquitin-mediated degradation of TßRI. HF disrupts the interaction between EPRS1 and TßRI, and reduces TßRI protein levels, leading to inhibition of the TGF-ß pathway. In conclusion, this work suggests the novel function of EPRS1 involved in the development of fibrosis by regulating the TGF-ß pathway and the antifibrotic effects of HF by controlling both of EPRS1 functions.


Subject(s)
Hepatic Stellate Cells , Receptors, Transforming Growth Factor beta , Humans , Hepatic Stellate Cells/metabolism , Receptor, Transforming Growth Factor-beta Type I/metabolism , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/metabolism , Fibrosis , Signal Transduction/physiology
8.
Adv Mater ; 35(19): e2206198, 2023 May.
Article in English | MEDLINE | ID: mdl-36856042

ABSTRACT

The sense of spiciness is related to the stimulation of vanilloid compounds contained in the foods. Although, the spiciness is commonly considered as the part of taste, it is more classified to the sense of pain stimulated on a tongue, namely, pungency, which is described as a tingling or burning on the tongue. Herein, first, a reusable electronic tongue based on a transient receptor potential vanilloid 1 (TRPV1) nanodisc conjugated graphene field-effect transistor is fabricated and spiciness-related pain evaluation with reusable electrode is demonstrated. The pungent compound reactive receptor TRPV1 is synthesized in the form of nanodiscs to maintain stability and reusability. The newly developed platform shows highly selective and sensitive performance toward each spiciness related vanilloid compound repeatably: 1 aM capsaicin, 10 aM dihydrocapsaicin, 1 fM piperine, 10 nM allicin, and 1 pM AITC. The binding mechanism is also examined by simulation. Furthermore, the elimination of the burning sensation on the tongue after eating spicy foods is not investigated. Based on the synthesis of micelles composed of casein protein (which is contained in skim milk) that remove pungent compounds bound to TRPV1 nanodisc, the deactivation of TRPV1 is investigated, and the electrode is reusable that mimics electronic tongue.


Subject(s)
Electronic Nose , Pain , Taste , Humans , Graphite
9.
J Bacteriol ; 194(14): 3749-50, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22740673

ABSTRACT

Enterohemorrhagic Escherichia coli causes severe food-borne disease in the guts of humans and animals. Here, we report the high-quality draft genome sequence of E. coli NCCP15658 isolated from a patient in the Republic of Korea. Its genome size was determined to be 5.46 Mb, and its genomic features, including genes encoding virulence factors, were analyzed.


Subject(s)
Enterohemorrhagic Escherichia coli/genetics , Genome, Bacterial , Enterohemorrhagic Escherichia coli/classification , Female , Gastrointestinal Diseases/epidemiology , Gastrointestinal Diseases/microbiology , Gene Expression Regulation, Bacterial/physiology , Humans , Molecular Sequence Data , Republic of Korea/epidemiology
10.
Nat Commun ; 13(1): 6455, 2022 10 29.
Article in English | MEDLINE | ID: mdl-36309524

ABSTRACT

The AKT signaling pathway plays critical roles in the resolution of inflammation. However, the underlying mechanisms of anti-inflammatory regulation and signal coordination remain unclear. Here, we report that anti-inflammatory AKT signaling is coordinated by glutamyl-prolyl-tRNA synthetase 1 (EPRS1). Upon inflammatory activation, AKT specifically phosphorylates Ser999 of EPRS1 in the cytoplasmic multi-tRNA synthetase complex, inducing release of EPRS1. EPRS1 compartmentalizes AKT to early endosomes via selective binding to the endosomal membrane lipid phosphatidylinositol 3-phosphate and assembles an AKT signaling complex specific for anti-inflammatory activity. These events promote AKT activation-mediated GSK3ß phosphorylation, which increase anti-inflammatory cytokine production. EPRS1-deficient macrophages do not assemble the early endosomal complex and consequently exacerbate inflammation, decreasing the survival of EPRS1-deficient mice undergoing septic shock and ulcerative colitis. Collectively, our findings show that the housekeeping protein EPRS1 acts as a mediator of inflammatory homeostasis by coordinating compartment-specific AKT signaling.


Subject(s)
Proto-Oncogene Proteins c-akt , Signal Transduction , Mice , Animals , Proto-Oncogene Proteins c-akt/metabolism , Anti-Inflammatory Agents/pharmacology , Inflammation
11.
J Immunother Cancer ; 8(1)2020 05.
Article in English | MEDLINE | ID: mdl-32461342

ABSTRACT

BACKGROUND: The generation of antigen-specific cytotoxic T lymphocyte (CTL) responses is required for successful cancer vaccine therapy. In this regard, ligands of Toll-like receptors (TLRs) have been suggested to activate adaptive immune responses by modulating the function of antigen-presenting cells (APCs). Despite their therapeutic potential, the development of TLR ligands for immunotherapy is often hampered due to rapid systemic toxicity. Regarding the safety concerns of currently available TLR ligands, finding a new TLR agonist with potent efficacy and safety is needed. METHODS: A unique structural domain (UNE-C1) was identified as a novel TLR2/6 in the catalytic region of human cysteinyl-tRNA synthetase 1 (CARS1) using comprehensive approaches, including RNA sequencing, the human embryonic kidney (HEK)-TLR Blue system, pull-down, and ELISA. The potency of its immunoadjuvant properties was analyzed by assessing antigen-specific antibody and CTL responses. In addition, the efficacy of tumor growth inhibition and the presence of the tumor-infiltrating leukocytes were evaluated using E.G7-OVA and TC-1 mouse models. The combined effect of UNE-C1 with an immune checkpoint inhibitor, anti-CTLA-4 antibody, was also evaluated in vivo. The safety of UNE-C1 immunization was determined by monitoring splenomegaly and cytokine production in the blood. RESULTS: Here, we report that CARS1 can be secreted from cancer cells to activate immune responses via specific interactions with TLR2/6 of APCs. A unique domain (UNE-C1) inserted into the catalytic region of CARS1 was determined to activate dendritic cells, leading to the stimulation of robust humoral and cellular immune responses in vivo. UNE-C1 also showed synergistic efficacy with cancer antigens and checkpoint inhibitors against different cancer models in vivo. Further, the safety assessment of UNE-C1 showed lower systemic cytokine levels than other known TLR agonists. CONCLUSIONS: We identified the endogenous TLR2/6 activating domain from human cysteinyl-tRNA synthetase CARS1. This novel TLR2/6 ligand showed potent immune-stimulating activity with little toxicity. Thus, the UNE-C1 domain can be developed as an effective immunoadjuvant with checkpoint inhibitors or cancer antigens to boost antitumor immunity.


Subject(s)
Amino Acyl-tRNA Synthetases/metabolism , Cancer Vaccines/administration & dosage , Immunity, Cellular/immunology , Immunotherapy/methods , Neoplasms, Experimental/therapy , Toll-Like Receptor 2/immunology , Amino Acyl-tRNA Synthetases/chemistry , Amino Acyl-tRNA Synthetases/immunology , Animals , Cancer Vaccines/immunology , Catalytic Domain , Dendritic Cells/immunology , Female , Humans , Immunization , Ligands , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/immunology , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , T-Lymphocytes, Cytotoxic/immunology , Toll-Like Receptor 2/chemistry , Toll-Like Receptor 2/metabolism
12.
mBio ; 9(1)2018 01 30.
Article in English | MEDLINE | ID: mdl-29382732

ABSTRACT

Pathogenic Vibrio species cause diseases in diverse marine animals reared in aquaculture. Since their pathogenesis, persistence, and survival in marine environments are regulated by quorum sensing (QS), QS interference has attracted attention as a means to control these bacteria in aquatic settings. A few QS inhibitors of Vibrio species have been reported, but detailed molecular mechanisms are lacking. Here, we identified a novel, potent, and selective Vibrio QS inhibitor, named QStatin [1-(5-bromothiophene-2-sulfonyl)-1H-pyrazole], which affects Vibrio harveyi LuxR homologues, the well-conserved master transcriptional regulators for QS in Vibrio species. Crystallographic and biochemical analyses showed that QStatin binds tightly to a putative ligand-binding pocket in SmcR, the LuxR homologue in V. vulnificus, and changes the flexibility of the protein, thereby altering its transcription regulatory activity. Transcriptome analysis revealed that QStatin results in SmcR dysfunction, affecting the expression of SmcR regulon required for virulence, motility/chemotaxis, and biofilm dynamics. Notably, QStatin attenuated representative QS-regulated phenotypes in various Vibrio species, including virulence against the brine shrimp (Artemia franciscana). Together, these results provide molecular insights into the mechanism of action of an effective, sustainable QS inhibitor that is less susceptible to resistance than other antimicrobial agents and useful in controlling the virulence of Vibrio species in aquacultures.IMPORTANCE Yields of aquaculture, such as penaeid shrimp hatcheries, are greatly affected by vibriosis, a disease caused by pathogenic Vibrio infections. Since bacterial cell-to-cell communication, known as quorum sensing (QS), regulates pathogenesis of Vibrio species in marine environments, QS inhibitors have attracted attention as alternatives to conventional antibiotics in aquatic settings. Here, we used target-based high-throughput screening to identify QStatin, a potent and selective inhibitor of V. harveyi LuxR homologues, which are well-conserved master QS regulators in Vibrio species. Structural and biochemical analyses revealed that QStatin binds tightly to a putative ligand-binding pocket on SmcR, the LuxR homologue in V. vulnificus, and affects expression of QS-regulated genes. Remarkably, QStatin attenuated diverse QS-regulated phenotypes in various Vibrio species, including pathogenesis against brine shrimp, with no impact on bacterial viability. Taken together, the results suggest that QStatin may be a sustainable antivibriosis agent useful in aquacultures.


Subject(s)
Anti-Bacterial Agents/pharmacology , Gene Expression Regulation, Bacterial/drug effects , Quorum Sensing/drug effects , Repressor Proteins/antagonists & inhibitors , Trans-Activators/antagonists & inhibitors , Vibrio/drug effects , Crystallography, X-Ray , Gene Expression Profiling , Protein Binding , Repressor Proteins/chemistry , Trans-Activators/chemistry
13.
Cancer Res ; 76(11): 3422-36, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27197155

ABSTRACT

AIMP2/p38 is a multifunctional tumor suppressor that normally resides in the cytosol as a scaffold protein of the multi-tRNA synthetase complex (MSC). One of the tumor-suppressive functions of AIMP2 is to facilitate ubiquitin-mediated degradation of FUSE-binding protein (FBP, FUBP1), a transcriptional activator of c-Myc. However, the mechanism by which AIMP2 functions within this pathway and its significance in tumorigenesis are uncertain. Here, we report that Smurf2 is responsible for AIMP2-mediated ubiquitination of FBP, and a mutation in AIMP2 that inhibited its nuclear interaction with Smurf2 enhanced cellular transformation and tumorigenesis in vivo Treatment of HeLa cells with TGFß resulted in the phosphorylation of AIMP2 on S156, a residue that is exposed on the embedded GST domain of AIMP2. We further found that phospho-AIMP2 dissociated from the MSC and translocated to the nucleus, where it bound to Smurf2, enhancing ubiquitination of FBP. AIMP2 also inhibited nuclear export of Smurf2 to sustain TGFß signaling. Collectively, these findings present a novel tumor-suppressive interaction between AIMP2 and Smurf2 and suggest that the disruption of this interaction can lead to oncogenic transformation. Cancer Res; 76(11); 3422-36. ©2016 AACR.


Subject(s)
Embryo, Mammalian/pathology , Fibroblasts/pathology , Lung Neoplasms/pathology , Peptide Elongation Factors/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Apoptosis , Blotting, Western , Cell Proliferation , Cells, Cultured , DNA Helicases/genetics , DNA Helicases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryo, Mammalian/metabolism , Female , Fibroblasts/metabolism , HeLa Cells , Humans , Immunoenzyme Techniques , Immunoprecipitation , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Nude , Peptide Elongation Factors/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/genetics , RNA-Binding Proteins , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Proteins/genetics , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination , Xenograft Model Antitumor Assays , p38 Mitogen-Activated Protein Kinases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL