Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Nat Immunol ; 17(4): 397-405, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26928339

ABSTRACT

The signaling adaptor TRAF3 is a highly versatile regulator of both innate immunity and adaptive immunity, but how its phosphorylation is regulated is still unknown. Here we report that deficiency in or inhibition of the conserved serine-threonine kinase CK1ɛ suppressed the production of type I interferon in response to viral infection. CK1ɛ interacted with and phosphorylated TRAF3 at Ser349, which thereby promoted the Lys63 (K63)-linked ubiquitination of TRAF3 and subsequent recruitment of the kinase TBK1 to TRAF3. Consequently, CK1ɛ-deficient mice were more susceptible to viral infection. Our findings establish CK1ɛ as a regulator of antiviral innate immune responses and indicate a novel mechanism of immunoregulation that involves CK1ɛ-mediated phosphorylation of TRAF3.


Subject(s)
Casein Kinase 1 epsilon/immunology , Immunity, Innate/immunology , Interferon-beta/immunology , TNF Receptor-Associated Factor 3/immunology , Animals , Casein Kinase 1 epsilon/antagonists & inhibitors , Casein Kinase 1 epsilon/genetics , Enzyme-Linked Immunosorbent Assay , HEK293 Cells , HeLa Cells , Herpes Simplex/immunology , Herpesvirus 1, Human/immunology , Humans , Interferon Type I/biosynthesis , Interferon Type I/immunology , Interferon-beta/biosynthesis , Mass Spectrometry , Mice , Mice, Knockout , Phosphorylation , Protein Serine-Threonine Kinases , Real-Time Polymerase Chain Reaction , Rhabdoviridae Infections/immunology , TNF Receptor-Associated Factor 3/genetics , Ubiquitination , Vesiculovirus/immunology , West Nile Fever/immunology , West Nile virus/immunology
2.
Nat Immunol ; 13(11): 1063-71, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23001144

ABSTRACT

The protein Tir (translocated intimin receptor) in enteric bacteria shares sequence similarity with the host cellular immunoreceptor tyrosine-based inhibition motifs (ITIMs). Despite the importance of Tir in pedestal formation, relatively little is known about the role of Tir and its ITIMs in the regulation of the host immune response. Here we demonstrate that Tir from enteropathogenic Escherichia coli (EPEC) interacted with the host cellular tyrosine phosphatase SHP-1 in an ITIM phosphorylation-dependent manner. The association of Tir with SHP-1 facilitated the recruitment of SHP-1 to the adaptor TRAF6 and inhibited the ubiquitination of TRAF6. Moreover, the ITIMs of Tir suppressed EPEC-stimulated expression of proinflammatory cytokines and inhibited intestinal immunity to infection with Citrobacter rodentium. Our findings identify a previously unknown mechanism by which bacterial ITIM-containing proteins can inhibit innate immune responses.


Subject(s)
Enterobacteriaceae Infections/immunology , Escherichia coli Proteins/immunology , Immunoreceptor Tyrosine-Based Inhibition Motif/immunology , Intestines/immunology , Receptors, Cell Surface/immunology , Signal Transduction/immunology , Animals , Cells, Cultured , Citrobacter rodentium/immunology , Enterobacteriaceae Infections/genetics , Enterobacteriaceae Infections/microbiology , Enteropathogenic Escherichia coli/immunology , Enteropathogenic Escherichia coli/pathogenicity , Escherichia coli Proteins/genetics , Gene Expression Regulation/immunology , Host-Pathogen Interactions , Immunity, Innate , Immunity, Mucosal , Immunoreceptor Tyrosine-Based Inhibition Motif/genetics , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestines/microbiology , Macrophages/immunology , Macrophages/microbiology , Mice , Phosphorylation , Protein Binding , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/immunology , Receptors, Cell Surface/genetics , Signal Transduction/genetics , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/immunology , Ubiquitin/genetics , Ubiquitin/immunology , Ubiquitination
3.
Opt Express ; 32(6): 8862-8876, 2024 Mar 11.
Article in English | MEDLINE | ID: mdl-38571133

ABSTRACT

An LP11-mode output all-fiber laser was presented, utilizing long-period fiber gratings (LPFGs) and polarization-maintaining optical fiber (PMF). The LPFG was designed and fabricated, achieving a 90.56% efficiency in LP01 to LP11 mode conversion. Furthermore, the transmission stability of LP11-mode in the PMF was also explored, with the spatial mode overlap ratio exceeding 0.95. Ultimately, the high-power polarization-maintaining (PM) fiber laser, capable of the LP11 mode output, was constructed, with the output power of 600 W and the beam quality M2 of 2.84. During the process of welding a thick Al-plate, the LP11 fiber laser exhibits a notable 1.88 times greater depth of fusion compared to the commercial single-mode fiber laser, when operating at the laser welding head speed of 100 mm/s. For applications demanding non-circular symmetric high-order modes, this research holds substantial potential for widespread adoption within the field of industrial processing.

4.
Nature ; 563(7729): 131-136, 2018 11.
Article in English | MEDLINE | ID: mdl-30356214

ABSTRACT

Accurate repair of DNA double-stranded breaks by homologous recombination preserves genome integrity and inhibits tumorigenesis. Cyclic GMP-AMP synthase (cGAS) is a cytosolic DNA sensor that activates innate immunity by initiating the STING-IRF3-type I IFN signalling cascade1,2. Recognition of ruptured micronuclei by cGAS links genome instability to the innate immune response3,4, but the potential involvement of cGAS in DNA repair remains unknown. Here we demonstrate that cGAS inhibits homologous recombination in mouse and human models. DNA damage induces nuclear translocation of cGAS in a manner that is dependent on importin-α, and the phosphorylation of cGAS at tyrosine 215-mediated by B-lymphoid tyrosine kinase-facilitates the cytosolic retention of cGAS. In the nucleus, cGAS is recruited to double-stranded breaks and interacts with PARP1 via poly(ADP-ribose). The cGAS-PARP1 interaction impedes the formation of the PARP1-Timeless complex, and thereby suppresses homologous recombination. We show that knockdown of cGAS suppresses DNA damage and inhibits tumour growth both in vitro and in vivo. We conclude that nuclear cGAS suppresses homologous-recombination-mediated repair and promotes tumour growth, and that cGAS therefore represents a potential target for cancer prevention and therapy.


Subject(s)
Cell Nucleus/metabolism , Cell Transformation, Neoplastic/pathology , Neoplasms/metabolism , Neoplasms/pathology , Nucleotidyltransferases/metabolism , Recombinational DNA Repair , Active Transport, Cell Nucleus , Adult , Animals , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/enzymology , DNA Breaks, Double-Stranded , DNA Damage , Female , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Neoplasm Proteins/metabolism , Neoplasms/genetics , Nucleotidyltransferases/deficiency , Phosphorylation , Phthalazines/pharmacology , Piperazines/pharmacology , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly (ADP-Ribose) Polymerase-1/metabolism , Protein Binding/drug effects , Recombinational DNA Repair/genetics , src-Family Kinases/metabolism
5.
Proc Natl Acad Sci U S A ; 118(16)2021 04 20.
Article in English | MEDLINE | ID: mdl-33846258

ABSTRACT

Macrophages are the key regulator of T-cell responses depending on their activation state. C-C motif chemokine receptor-like 2 (CCRL2), a nonsignaling atypical receptor originally cloned from LPS-activated macrophages, has recently been shown to regulate immune responses under several inflammatory conditions. However, whether CCRL2 influences macrophage function and regulates tumor immunity remains unknown. Here, we found that tumoral CCRL2 expression is a predictive indicator of robust antitumor T-cell responses in human cancers. CCRL2 is selectively expressed in tumor-associated macrophages (TAM) with immunostimulatory phenotype in humans and mice. Conditioned media from tumor cells could induce CCRL2 expression in macrophages primarily via TLR4, which is negated by immunosuppressive factors. Ccrl2-/- mice exhibit accelerated melanoma growth and impaired antitumor immunity characterized by significant reductions in immunostimulatory macrophages and T-cell responses in tumor. Depletion of CD8+ T cells or macrophages eliminates the difference in tumor growth between WT and Ccrl2-/- mice. Moreover, CCRL2 deficiency impairs immunogenic activation of macrophages, resulting in attenuated antitumor T-cell responses and aggravated tumor growth in a coinjection tumor model. Mechanically, CCRL2 interacts with TLR4 on the cell surface to retain membrane TLR4 expression and further enhance its downstream Myd88-NF-κB inflammatory signaling in macrophages. Similarly, Tlr4-/- mice exhibit reduced CCRL2 expression in TAM and accelerated melanoma growth. Collectively, our study reveals a functional role of CCRL2 in activating immunostimulatory macrophages, thereby potentiating antitumor T-cell response and tumor rejection, and suggests CCLR2 as a potential biomarker candidate and therapeutic target for cancer immunotherapy.


Subject(s)
Macrophage Activation/immunology , Neoplasms/immunology , Receptors, CCR/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , China , Female , Immunization , Macrophage Activation/physiology , Male , Melanoma/metabolism , Mice , NF-kappa B/metabolism , Neoplasms/genetics , Receptors, CCR/genetics , Signal Transduction , T-Lymphocytes/metabolism , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism
6.
J Immunol ; 206(6): 1161-1170, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33568397

ABSTRACT

Helicobacter pylori is the major etiological agent for most gastric cancer. CagA has been reported to be an important virulence factor of H. pylori, but its effect on the immune response is not yet clear. In this study, wild-type C57BL/6 mice and Ptpn6me-v/me-v mice were randomly assigned for infection with H. pylori We demonstrated that CagA suppressed H. pylori-stimulated expression of proinflammatory cytokines in vivo. Besides, we infected mouse peritoneal macrophages RAW264.7 and AGS with H. pylori Our results showed that CagA suppressed expression of proinflammatory cytokines through inhibiting the MAPKs and NF-κB pathways activation in vitro. Mechanistically, we found that CagA interacted with the host cellular tyrosine phosphatase SHP-1, which facilitated the recruitment of SHP-1 to TRAF6 and inhibited the K63-linked ubiquitination of TRAF6, which obstructed the transmission of signal downstream. Taken together, these findings reveal a previously unknown mechanism by which CagA negatively regulates the posttranslational modification of TRAF6 in innate antibacterial immune response and provide molecular basis for new therapeutics to treat microbial infection.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Helicobacter Infections/immunology , Helicobacter pylori/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , TNF Receptor-Associated Factor 6/metabolism , Animals , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Disease Models, Animal , Gastric Mucosa/immunology , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , HEK293 Cells , HeLa Cells , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter pylori/metabolism , Humans , Immunity, Innate , Lysine/metabolism , Macrophages, Peritoneal , Male , Mice , Mice, Transgenic , Primary Cell Culture , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , RAW 264.7 Cells , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction/immunology , Transfection , Ubiquitination/immunology
7.
FASEB J ; 34(9): 12392-12405, 2020 09.
Article in English | MEDLINE | ID: mdl-32779804

ABSTRACT

Type I interferons play a pivotal role in innate immune response to virus infection. The protein tyrosine phosphatase SHP-1 was reported to function as a negative regulator of inflammatory cytokine production by inhibiting activation of NF-κB and MAPKs during bacterial infection, however, the role of SHP-1 in regulating type I interferons remains unknown. Here, we demonstrated that knockout or knockdown of SHP-1 in macrophages promoted both HSV-1- and VSV-induced antiviral immune response. Conversely, overexpression of SHP-1 in L929 cells suppressed the HSV-1- and VSV-induced immune response; suppression was directly dependent on phosphatase activity. We identified a direct interaction between SHP-1 and TRAF3; the association between these two proteins resulted in diminished recruitment of CK1ε to TRAF3 and inhibited its K63-linked ubiquitination; SHP-1 inhibited K63-linked ubiquitination of TRAF3 by promoting dephosphorylation at Tyr116 and Tyr446. Taken together, our results identify SHP-1 as a negative regulator of antiviral immunity and suggest that SHP-1 may be a target for intervention in acute virus infection.


Subject(s)
Protein Tyrosine Phosphatase, Non-Receptor Type 6/physiology , TNF Receptor-Associated Factor 3/physiology , Virus Diseases/immunology , Animals , HEK293 Cells , Humans , Immunity, Innate , Mice , RAW 264.7 Cells , Ubiquitination
8.
PLoS Pathog ; 13(6): e1006436, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28622363

ABSTRACT

Pulmonary infection is the most common risk factor for acute lung injury (ALI). Innate immune responses induced by Microbe-Associated Molecular Pattern (MAMP) molecules are essential for lung defense but can lead to tissue injury. Little is known about how MAMP molecules are degraded in the lung or how MAMP degradation/inactivation helps prevent or ameliorate the harmful inflammation that produces ALI. Acyloxyacyl hydrolase (AOAH) is a host lipase that inactivates Gram-negative bacterial endotoxin (lipopolysaccharide, or LPS). We report here that alveolar macrophages increase AOAH expression upon exposure to LPS and that Aoah+/+ mice recover more rapidly than do Aoah-/- mice from ALI induced by nasally instilled LPS or Klebsiella pneumoniae. Aoah-/- mouse lungs had more prolonged leukocyte infiltration, greater pro- and anti-inflammatory cytokine expression, and longer-lasting alveolar barrier damage. We also describe evidence that the persistently bioactive LPS in Aoah-/- alveoli can stimulate alveolar macrophages directly and epithelial cells indirectly to produce chemoattractants that recruit neutrophils to the lung and may prevent their clearance. Distinct from the prolonged tolerance observed in LPS-exposed Aoah-/- peritoneal macrophages, alveolar macrophages that lacked AOAH maintained or increased their responses to bioactive LPS and sustained inflammation. Inactivation of LPS by AOAH is a previously unappreciated mechanism for promoting resolution of pulmonary inflammation/injury induced by Gram-negative bacterial infection.


Subject(s)
Acute Lung Injury/immunology , Carboxylic Ester Hydrolases/immunology , Lipopolysaccharides/adverse effects , Acute Lung Injury/enzymology , Acute Lung Injury/etiology , Animals , Carboxylic Ester Hydrolases/genetics , Humans , Klebsiella Infections/enzymology , Klebsiella Infections/genetics , Klebsiella Infections/immunology , Klebsiella pneumoniae/immunology , Lipopolysaccharides/immunology , Lung/immunology , Lung/microbiology , Macrophages, Peritoneal/enzymology , Macrophages, Peritoneal/immunology , Mice , Mice, Knockout
9.
J Infect Dis ; 218(2): 312-323, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29228365

ABSTRACT

Tuberculosis, caused by Mycobacterium tuberculosis infection, remains a global threat to human health, but knowledge of the molecular mechanisms underlying the pathogenesis of tuberculosis is still limited. Although Notch4, a member of the Notch receptor family, is involved in the initiation of mammary tumors, its function in M. tuberculosis infection remains unclear. In this study, we found that Notch4-deficient mice were more resistant to M. tuberculosis infection, with a much lower bacterial burden and fewer pathological changes in the lungs. Notch4 inhibited M. tuberculosis-induced production of proinflammatory cytokines by interaction with TAK1 and inhibition of its activation. Furthermore, we found that Notch intracellular domain 4 prevented TRAF6 autoubiquitination and suppressed TRAF6-mediated TAK1 polyubiquitination. Finally, Notch inhibitors made mice more resistant to M. tuberculosis infection. These results suggest that Notch4 is a negative regulator of M. tuberculosis-induced inflammatory response, and treatment with a Notch inhibitor could serve as a new therapeutic strategy for tuberculosis.


Subject(s)
Gene Expression Regulation , MAP Kinase Kinase Kinases/metabolism , Receptor, Notch4/metabolism , Tuberculosis, Pulmonary/pathology , Animals , Bacterial Load , Cytokines/analysis , Disease Models, Animal , Female , HEK293 Cells , Humans , Inflammation/pathology , Lung/microbiology , Lung/pathology , Male , Mice , Mice, Inbred C57BL , RAW 264.7 Cells , Receptor, Notch4/deficiency , TNF Receptor-Associated Factor 6/metabolism , Tuberculosis, Pulmonary/microbiology
10.
Clin Immunol ; 194: 1-8, 2018 09.
Article in English | MEDLINE | ID: mdl-29906512

ABSTRACT

Increasing rates of life-threatening infections and decreasing susceptibility to antibiotics urge an effective vaccine targeting Staphylococcus aureus. Here we investigate the role of cellular immunity in FnBPA110-263 mediated protection in Staphylococcus aureus infection. This study revealed FnBPA110-263 broadly protected mice from seven FnBPA isotypes strains in the sepsis model. FnBPA110-263 immunized B-cell deficient mice were protected against lethal challenge, while T-cell deficient mice were not. Reconstituting mice with FnBPA110-263 specific CD4+ T-cells conferred antigen specific protection. In vitro assays indicated that isolated FnBPA110-263 specific splenocytes from immunized mice produced abundant IL-17A. IL-17A deficient mice were not protected from a lethal challenge by FnBPA110-263 vaccination. Moreover, neutralizing IL-17A, but not IFN-γ,reverses FnBPA110-263-induced protective efficacy in sepsis and skin infection model. These findings suggest that IL-17A producing Th17 cells play an essential role in FnBPA110-263 vaccine-mediated defense against S. aureus sepsis and skin infection in mice.


Subject(s)
Adhesins, Bacterial/immunology , Bacterial Vaccines/immunology , Sepsis/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Animals , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/microbiology , Immunity, Cellular/immunology , Interferon-gamma/immunology , Interleukin-17/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Mice, SCID , Sepsis/microbiology , Th17 Cells/immunology , Th17 Cells/microbiology , Vaccination/methods
11.
Proc Natl Acad Sci U S A ; 112(48): 14942-7, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26627244

ABSTRACT

FoxP3(+) T regulatory (Treg) cells have a fundamental role in immunological tolerance, with transcriptional and functional phenotypes that demarcate them from conventional CD4(+) T cells (Tconv). Differences between these two lineages in the signaling downstream of T-cell receptor-triggered activation have been reported, and there are different requirements for some signaling factors. Seeking a comprehensive view, we found that Treg cells have a broadly dampened activation of several pathways and signaling nodes upon TCR-mediated activation, with low phosphorylation of CD3ζ, SLP76, Erk1/2, AKT, or S6 and lower calcium flux. In contrast, STAT phosphorylation triggered by interferons, IL2 or IL6, showed variations between Treg and Tconv in magnitude or choice of preferential STAT activation but no general Treg signaling defect. Much, but not all, of the Treg/Tconv difference in TCR-triggered responses could be attributed to lower responsiveness of antigen-experienced cells with CD44(hi) or CD62L(lo) phenotypes, which form a greater proportion of the Treg pool. Candidate regulators were tested, but the Treg/Tconv differential could not be explained by overexpression in Treg cells of the signaling modulator CD5, the coinhibitors PD-1 and CTLA4, or the regulatory phosphatase DUSP4. However, transcriptome profiling in Dusp4-deficient mice showed that DUSP4 enhances the expression of a segment of the canonical Treg transcriptional signature, which partially overlaps with the TCR-dependent Treg gene set. Thus, Treg cells, likely because of their intrinsically higher reactivity to self, tune down TCR signals but seem comparatively more attuned to cytokines or other intercellular signals.


Subject(s)
Calcium Signaling/immunology , Forkhead Transcription Factors/immunology , Gene Expression Regulation/immunology , MAP Kinase Signaling System/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens, CD/genetics , Antigens, CD/immunology , CTLA-4 Antigen/genetics , CTLA-4 Antigen/immunology , Calcium Signaling/genetics , Forkhead Transcription Factors/genetics , MAP Kinase Signaling System/genetics , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/immunology , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/immunology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology
12.
Adv Exp Med Biol ; 1024: 37-61, 2017.
Article in English | MEDLINE | ID: mdl-28921464

ABSTRACT

Inflammation is usually the defensive reaction of the immune system to the invasion of pathogen and the exogenous objects. The activation of inflammation helps our body to eliminate pathogenic microbe, virus, and parasite harming our health, while under many circumstances inflammation is the direct cause of the pathological damage in tissues and dysfunction of organs. The posttranslational modification (PTM) of the inflammatory pathways, such as TLR pathways, RLR pathways, NLR pathway, intracellular DNA sensors, intracellular RNA sensors, and inflammasomes, is crucial in the regulation of these signaling trails. Ubiquitination, phosphorylation, polyubiquitination, methylation, and acetylation are the main forms of the PTM, and they respectively play different roles in signaling regulation. The effects of the PTM range from the production of pro-inflammatory factors and the interaction between adaptors and receptors to cell translocation in response to the infectious or other dangerous factors. In this chapter, we will have an overview of the different ways of the posttranslational modifications in different inflammatory signaling pathways and their essential roles in regulation of inflammation.


Subject(s)
Gene Expression Regulation/immunology , Inflammation/metabolism , Protein Processing, Post-Translational/immunology , Signal Transduction/immunology , Animals , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism , Gene Expression Regulation/physiology , Humans , Inflammasomes , Inflammation/immunology , NLR Proteins/genetics , NLR Proteins/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Phosphorylation , RNA/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Toll-Like Receptors/immunology , Ubiquitination
13.
Opt Express ; 24(13): 14463-9, 2016 Jun 27.
Article in English | MEDLINE | ID: mdl-27410599

ABSTRACT

We report a high power all-fiber amplifier with suitable seed power injected by an all-fiber laser. Different seed powers were injected into the all-fiber amplifier during our amplification experiments, and we found the stimulated Raman scattering (SRS) threshold was inversely proportional to the injected seed power. More than 3 kW signal light with good beam quality (M2 = 1.28) has been obtained with a suitable seed power injected, and the slope efficiency of the all-fiber amplifier was about 84.4%.

14.
Front Immunol ; 14: 1211816, 2023.
Article in English | MEDLINE | ID: mdl-37854611

ABSTRACT

SARS-COV-2 infection-induced excessive or uncontrolled cytokine storm may cause injury of host tissue or even death. However, the mechanism by which SARS-COV-2 causes the cytokine storm is unknown. Here, we demonstrated that SARS-COV-2 protein NSP9 promoted cytokine production by interacting with and activating TANK-binding kinase-1 (TBK1). With an rVSV-NSP9 virus infection model, we discovered that an NSP9-induced cytokine storm exacerbated tissue damage and death in mice. Mechanistically, NSP9 promoted the K63-linked ubiquitination and phosphorylation of TBK1, which induced the activation and translocation of IRF3, thereby increasing downstream cytokine production. Moreover, the E3 ubiquitin ligase Midline 1 (MID1) facilitated the K48-linked ubiquitination and degradation of NSP9, whereas virus infection inhibited the interaction between MID1 and NSP9, thereby inhibiting NSP9 degradation. Additionally, we identified Lys59 of NSP9 as a critical ubiquitin site involved in the degradation. These findings elucidate a previously unknown mechanism by which a SARS-COV-2 protein promotes cytokine storm and identifies a novel target for COVID-19 treatment.


Subject(s)
COVID-19 , Cytokine Release Syndrome , Protein Serine-Threonine Kinases , SARS-CoV-2 , Animals , Mice , COVID-19/complications , COVID-19/genetics , COVID-19/immunology , COVID-19 Drug Treatment , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/genetics , Cytokine Release Syndrome/immunology , Cytokines , Disease Models, Animal , Immunity, Innate , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , SARS-CoV-2/genetics , SARS-CoV-2/immunology , SARS-CoV-2/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/metabolism
15.
Cell Rep ; 42(5): 112442, 2023 05 30.
Article in English | MEDLINE | ID: mdl-37099423

ABSTRACT

Cyclic GMP-AMP synthase (cGAS) recognizes Y-form cDNA of human immunodeficiency virus type 1 (HIV-1) and initiates antiviral immune response through cGAS-stimulator of interferon genes (STING)-TBK1-IRF3-type I interferon (IFN-I) signalingcascade. Here, we report that the HIV-1 p6 protein suppresses HIV-1-stimulated expression of IFN-I and promotes immune evasion. Mechanistically, the glutamylated p6 at residue Glu6 inhibits the interaction between STING and tripartite motif protein 32 (TRIM32) or autocrine motility factor receptor (AMFR). This subsequently suppresses the K27- and K63-linked polyubiquitination of STING at K337, therefore inhibiting STING activation, whereas mutation of the Glu6 residue partially reverses the inhibitory effect. However, CoCl2, an agonist of cytosolic carboxypeptidases (CCPs), counteracts the glutamylation of p6 at the Glu6 residue and inhibits HIV-1 immune evasion. These findings reveal a mechanism through which an HIV-1 protein mediates immune evasion and provides a therapeutic drug candidate to treat HIV-1 infection.


Subject(s)
HIV-1 , Humans , HIV-1/metabolism , Signal Transduction , Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism , Immunity, Innate/genetics
16.
Cell Mol Immunol ; 19(1): 108-121, 2022 01.
Article in English | MEDLINE | ID: mdl-34811497

ABSTRACT

HIV-1 infection-induced cGAS-STING-TBK1-IRF3 signaling activates innate immunity to produce type I interferon (IFN). The HIV-1 nonstructural protein viral infectivity factor (Vif) is essential in HIV-1 replication, as it degrades the host restriction factor APOBEC3G. However, whether and how it regulates the host immune response remains to be determined. In this study, we found that Vif inhibited the production of type I IFN to promote immune evasion. HIV-1 infection induced the activation of the host tyrosine kinase FRK, which subsequently phosphorylated the immunoreceptor tyrosine-based inhibitory motif (ITIM) of Vif and enhanced the interaction between Vif and the cellular tyrosine phosphatase SHP-1 to inhibit type I IFN. Mechanistically, the association of Vif with SHP-1 facilitated SHP-1 recruitment to STING and inhibited the K63-linked ubiquitination of STING at Lys337 by dephosphorylating STING at Tyr162. However, the FRK inhibitor D-65495 counteracted the phosphorylation of Vif to block the immune evasion of HIV-1 and antagonize infection. These findings reveal a previously unknown mechanism through which HIV-1 evades antiviral immunity via the ITIM-containing protein to inhibit the posttranslational modification of STING. These results provide a molecular basis for the development of new therapeutic strategies to treat HIV-1 infection.


Subject(s)
HIV-1 , Antiviral Agents , Immune Evasion , Immunity, Innate , Protein Serine-Threonine Kinases
17.
Nat Commun ; 13(1): 5493, 2022 09 19.
Article in English | MEDLINE | ID: mdl-36123338

ABSTRACT

Many pathogens secrete effectors to hijack intracellular signaling regulators in host immune cells to promote pathogenesis. However, the pathogenesis of Staphylococcus aureus secretory effectors within host cells is unclear. Here, we report that Staphylococcus aureus secretes extracellular fibrinogen-binding protein (Efb) into the cytoplasm of macrophages to suppress host immunity. Mechanistically, RING finger protein 114, a host E3 ligase, mediates K27-linked ubiquitination of Efb at lysine 71, which facilitates the recruitment of tumor necrosis factor receptor associated factor (TRAF) 3. The binding of Efb to TRAF3 disrupts the formation of the TRAF3/TRAF2/cIAP1 (cellular-inhibitor-of-apoptosis-1) complex, which mediates K48-ubiquitination of TRAF3 to promote degradation, resulting in suppression of the inflammatory signaling cascade. Additionally, the Efb K71R mutant loses the ability to inhibit inflammation and exhibits decreased pathogenicity. Therefore, our findings identify an unrecognized mechanism of Staphylococcus aureus to suppress host defense, which may be a promising target for developing effective anti-Staphylococcus aureus immunomodulators.


Subject(s)
Staphylococcal Infections , TNF Receptor-Associated Factor 3 , Fibrinogen/metabolism , Humans , Lysine/metabolism , Staphylococcus aureus/metabolism , TNF Receptor-Associated Factor 2/metabolism , TNF Receptor-Associated Factor 3/metabolism , Ubiquitin-Protein Ligases/metabolism
18.
Nat Commun ; 11(1): 6000, 2020 11 26.
Article in English | MEDLINE | ID: mdl-33243993

ABSTRACT

Virus infection may induce excessive interferon (IFN) responses that can lead to host tissue injury or even death. ß-arrestin 2 regulates multiple cellular events through the G protein-coupled receptor (GPCR) signaling pathways. Here we demonstrate that ß-arrestin 2 also promotes virus-induced production of IFN-ß and clearance of viruses in macrophages. ß-arrestin 2 interacts with cyclic GMP-AMP synthase (cGAS) and increases the binding of dsDNA to cGAS to enhance cyclic GMP-AMP (cGAMP) production and the downstream stimulator of interferon genes (STING) and innate immune responses. Mechanistically, deacetylation of ß-arrestin 2 at Lys171 facilitates the activation of the cGAS-STING signaling and the production of IFN-ß. In vitro, viral infection induces the degradation of ß-arrestin 2 to facilitate immune evasion, while a ß-blocker, carvedilol, rescues ß-arrestin 2 expression to maintain the antiviral immune response. Our results thus identify a viral immune-evasion pathway via the degradation of ß-arrestin 2, and also hint that carvedilol, approved for treating heart failure, can potentially be repurposed as an antiviral drug candidate.


Subject(s)
Carvedilol/pharmacology , Immune Evasion/immunology , Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism , Virus Diseases/immunology , beta-Arrestin 2/metabolism , Animals , Carvedilol/therapeutic use , Disease Models, Animal , Drug Repositioning , HEK293 Cells , Herpesvirus 1, Human/immunology , Humans , Immune Evasion/drug effects , Interferon-beta/metabolism , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Male , Mice , Primary Cell Culture , Proteolysis/drug effects , RAW 264.7 Cells , RNA-Seq , Sendai virus/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , Vesiculovirus/immunology , Virus Diseases/drug therapy , Virus Diseases/virology , beta-Arrestin 2/agonists , beta-Arrestin 2/genetics
19.
Gut Microbes ; 11(5): 1423-1437, 2020 09 02.
Article in English | MEDLINE | ID: mdl-32403971

ABSTRACT

The protein translocated intimin receptor (Tir) from enteropathogenic Escherichia coli shares sequence similarity with the host cellular immunoreceptor tyrosine-based inhibition motifs (ITIMs). The ITIMs of Tir are required for Tir-mediated immune inhibition and evasion of host immune responses. However, the underlying molecular mechanism by which Tir regulates immune inhibition remains unclear. Here we demonstrated that ß-arrestin 2, which is involved in the G-protein-coupled receptor (GPCR) signal pathway, interacted with Tir in an ITIM-dependent manner. For the molecular mechanism, we found that ß-arrestin 2 enhanced the recruitment of SHP-1 to Tir. The recruited SHP-1 inhibited K63-linked ubiquitination of TRAF6 by dephosphorylating TRAF6 at Tyr288, and inhibited K63-linked ubiquitination and phosphorylation of TAK1 by dephosphorylating TAK1 at Tyr206, which cut off the downstream signal transduction and subsequent cytokine production. Moreover, the inhibitory effect of Tir on immune responses was diminished in ß-arrestin 2-deficient mice and macrophages. These findings suggest that ß-arrestin 2 is a key regulator in Tir-mediated immune evasion, which could serve as a new therapeutic target for bacterial infectious diseases.


Subject(s)
Enteropathogenic Escherichia coli/pathogenicity , Immune Evasion , Macrophages/microbiology , Toll-Like Receptors/metabolism , beta-Arrestin 2/metabolism , Amino Acid Motifs , Animals , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Enteropathogenic Escherichia coli/immunology , Enteropathogenic Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , MAP Kinase Kinase Kinases/metabolism , Macrophages, Peritoneal/microbiology , Mice , Mice, Inbred C57BL , Phosphorylation , Protein Binding , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , RAW 264.7 Cells , RNA, Small Interfering , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism , Signal Transduction , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/metabolism , beta-Arrestin 2/genetics
20.
Cell Signal ; 20(11): 2002-12, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18713649

ABSTRACT

The adaptor protein paxillin plays an important role in cell migration. Although the c-Jun amino-terminal kinase (JNK) phosphorylation of paxillin on Ser 178 has been found to be critical for cell migration, the precise mechanism by which JNK regulates cell migration is still not very clear. Here, the migration of human corneal epithelial (HCE) cells was used to determine which signaling pathways are involved in EGF-induced paxillin phosphorylation. Paxillin was phosphorylated on Tyr 31 and Tyr 118 after induction of migration by EGF in HCE cells. Specific inhibition of JNK activation by inhibitor SP600125 or overexpression of a dominant-negative JNK mutant not only blocked EGF-induced cell migration, but also eliminated tyrosine phosphorylation of paxillin on Tyr 31 and Tyr 118. HCE cells overexpressing paxillin-S178A mutant also exhibited lower mobility, and reduced phosphorylation of Tyr 31 and Tyr 118. However, paxillin-S178A-inhibited cell migration can be rescued by overexpression of paxillin-Y31E/Y118E mutant. Importantly, inhibition of JNK by SP600125 or overexpression of paxillin-S178A mutant prevented the association of FAK with paxillin. Taken together, these results suggest that phosphorylation of paxillin on Ser 178 by JNK is required for the association of paxillin with FAK, and subsequent tyrosine phosphorylation of paxillin.


Subject(s)
Cell Movement , Epithelial Cells/cytology , Epithelium, Corneal/cytology , JNK Mitogen-Activated Protein Kinases/metabolism , Paxillin/metabolism , Phosphotyrosine/metabolism , Cell Line , Cell Movement/drug effects , Enzyme Activation/drug effects , Epidermal Growth Factor/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , MAP Kinase Signaling System/drug effects , Models, Biological , Mutant Proteins/metabolism , Phosphorylation/drug effects , Phosphoserine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL