Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 53(1): 35-46, 2024 Jan 19.
Article in English, Chinese | MEDLINE | ID: mdl-38426691

ABSTRACT

Innate nucleic acid sensing is a ubiquitous and highly conserved immunological process, which is pivotal for monitoring and responding to pathogenic invasion and cellular damage, and central to host defense, autoimmunity, cell fate determination and tumorigenesis. Tyrosine phosphorylation, a major type of post-translational modification, plays a critical regulatory role in innate immune sensing pathway. Core members of nucleic acid sensing signaling pathway, such as cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS), stimulator of interferon genes (STING), and TANK binding kinase 1 (TBK1), are all subject to activity regulation triggered by tyrosine phosphorylation, thereby affecting the host antiviral defense and anti-tumor immunity under physiological or pathological conditions. This review summarizes the recent advances in research on tyrosine kinases and tyrosine phosphorylation in regulation of nucleic acid sensing. The function and potential applications of targeting tyrosine phosphorylation in anti-tumor immunity is disussed to provide insights for understanding and expanding new anti-tumor strategies.


Subject(s)
Nucleic Acids , Protein-Tyrosine Kinases , Immunity, Innate , Signal Transduction , Tyrosine
2.
Sci Adv ; 10(9): eadj2102, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38416816

ABSTRACT

Cytosolic double-stranded DNA surveillance by cyclic GMP-AMP synthase (cGAS)-Stimulator of Interferon Genes (STING) signaling triggers cellular senescence, autophagy, biased mRNA translation, and interferon-mediated immune responses. However, detailed mechanisms and physiological relevance of STING-induced senescence are not fully understood. Here, we unexpectedly found that interferon regulatory factor 3 (IRF3), activated during innate DNA sensing, forms substantial endogenous complexes in the nucleus with retinoblastoma (RB), a key cell cycle regulator. The IRF3-RB interaction attenuates cyclin-dependent kinase 4/6 (CDK4/6)-mediated RB hyperphosphorylation that mobilizes RB to deactivate E2 family (E2F) transcription factors, thereby driving cells into senescence. STING-IRF3-RB signaling plays a notable role in hepatic stellate cells (HSCs) within various murine models, pushing activated HSCs toward senescence. Accordingly, IRF3 global knockout or conditional deletion in HSCs aggravated liver fibrosis, a process mitigated by the CDK4/6 inhibitor. These findings underscore a straightforward yet vital mechanism of cGAS-STING signaling in inducing cellular senescence and unveil its unexpected biology in limiting liver fibrosis.


Subject(s)
Retinal Neoplasms , Retinoblastoma , Mice , Animals , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , DNA/metabolism , Interferons/metabolism
3.
Nat Cell Biol ; 26(2): 219-234, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38253667

ABSTRACT

Lysosomal storage disorders (LSDs), which are characterized by genetic and metabolic lysosomal dysfunctions, constitute over 60 degenerative diseases with considerable health and economic burdens. However, the mechanisms driving the progressive death of functional cells due to lysosomal defects remain incompletely understood, and broad-spectrum therapeutics against LSDs are lacking. Here, we found that various gene abnormalities that cause LSDs, including Hexb, Gla, Npc1, Ctsd and Gba, all shared mutual properties to robustly autoactivate neuron-intrinsic cGAS-STING signalling, driving neuronal death and disease progression. This signalling was triggered by excessive cytoplasmic congregation of the dsDNA and DNA sensor cGAS in neurons. Genetic ablation of cGAS or STING, digestion of neuronal cytosolic dsDNA by DNase, and repair of neuronal lysosomal dysfunction alleviated symptoms of Sandhoff disease, Fabry disease and Niemann-Pick disease, with substantially reduced neuronal loss. We therefore identify a ubiquitous mechanism mediating the pathogenesis of a variety of LSDs, unveil an inherent connection between lysosomal defects and innate immunity, and suggest a uniform strategy for curing LSDs.


Subject(s)
Lysosomal Storage Diseases , Niemann-Pick Disease, Type C , Humans , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/metabolism , Lysosomal Storage Diseases/pathology , Niemann-Pick Disease, Type C/genetics , Niemann-Pick Disease, Type C/pathology , Lysosomes/metabolism , Immunity, Innate , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism
5.
J Hepatol ; 78(4): 704-716, 2023 04.
Article in English | MEDLINE | ID: mdl-36574921

ABSTRACT

BACKGROUND & AIMS: Appropriate treatment options are lacking for hepatitis E virus (HEV)-infected pregnant women and immunocompromised individuals. Thus, we aimed to identify efficient anti-HEV drugs through high-throughput screening, validate them in vitro and in vivo (in a preclinical animal study), and elucidate their underlying antiviral mechanism of action. METHODS: Using appropriate cellular and rodent HEV infection models, we studied a critical pathway for host-HEV interactions and performed a preclinical study of the corresponding antivirals, which target proteostasis of the HEV replicase. RESULTS: We found 17 inhibitors that target HEV-HSP90 interactions by unbiased compound library screening on human hepatocytes harboring an HEV replicon. Inhibitors of HSP90 (iHSP90) markedly suppressed HEV replication with efficacy exceeding that of conventional antivirals (IFNα and ribavirin) in vitro. Mechanistically, iHSP90 treatment released the viral replicase ORF1 protein from the ORF1-HSP90 complex and triggered rapid ubiquitin/proteasome-mediated degradation of ORF1, resulting in abrogated HEV replication. Furthermore, a preclinical trial in a Mongolian gerbil HEV infection model showed this novel anti-HEV strategy to be safe, efficient, and able to prevent HEV-induced liver damage. CONCLUSIONS: In this study, we uncover a proteostatic pathway that is critical for host-HEV interactions and we provide a foundation from which to translate this new understanding of the HEV life cycle into clinically promising antivirals. IMPACT AND IMPLICATIONS: Appropriate treatment options for hepatitis E virus (HEV)-infected pregnant women and immunocompromised patients are lacking; hence, there is an urgent need for safe and effective HEV-specific therapies. This study identified new antivirals (inhibitors of HSP90) that significantly limit HEV infection by targeting the viral replicase for degradation. Moreover, these anti-HEV drugs were validated in an HEV rodent model and were found to be safe and efficient for prevention of HEV-induced liver injury in preclinical experiments. Our findings substantially promote the understanding of HEV pathobiology and pave the way for antiviral development.


Subject(s)
Hepatitis E virus , Hepatitis E , Animals , Humans , Female , Pregnancy , Proteostasis , Viral Replicase Complex Proteins , Hepatitis E/drug therapy , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Viral Proteins , Virus Replication
6.
Mol Cell ; 82(23): 4519-4536.e7, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36384137

ABSTRACT

Nutrient sensing and damage sensing are two fundamental processes in living organisms. While hyperglycemia is frequently linked to diabetes-related vulnerability to microbial infection, how body glucose levels affect innate immune responses to microbial invasion is not fully understood. Here, we surprisingly found that viral infection led to a rapid and dramatic decrease in blood glucose levels in rodents, leading to robust AMPK activation. AMPK, once activated, directly phosphorylates TBK1 at S511, which triggers IRF3 recruitment and the assembly of MAVS or STING signalosomes. Consistently, ablation or inhibition of AMPK, knockin of TBK1-S511A, or increased glucose levels compromised nucleic acid sensing, while boosting AMPK-TBK1 cascade by AICAR or TBK1-S511E knockin improves antiviral immunity substantially in various animal models. Thus, we identify TBK1 as an AMPK substrate, reveal the molecular mechanism coupling a dual sensing of glucose and nuclei acids, and report its physiological necessity in antiviral defense.


Subject(s)
AMP-Activated Protein Kinases , Nucleic Acids , Animals , AMP-Activated Protein Kinases/genetics , Immunity, Innate , Antiviral Agents , Glucose
7.
Nat Cell Biol ; 24(5): 766-782, 2022 05.
Article in English | MEDLINE | ID: mdl-35501370

ABSTRACT

Innate DNA sensing via the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) mechanism surveys microbial invasion and cellular damage and thus participates in various human infectious diseases, autoimmune diseases and cancers. However, how DNA sensing rapidly and adaptively shapes cellular physiology is incompletely known. Here we identify the STING-PKR-like endoplasmic reticulum kinase (PERK)-eIF2α pathway, a previously unknown cGAS-STING mechanism, enabling an innate immunity control of cap-dependent messenger RNA translation. Upon cGAMP binding, STING at the ER binds and directly activates the ER-located kinase PERK via their intracellular domains, which precedes TBK1-IRF3 activation and is irrelevant to the unfolded protein response. The activated PERK phosphorylates eIF2α, forming an inflammatory- and survival-preferred translation program. Notably, this STING-PERK-eIF2α pathway is evolutionarily primitive and physiologically critical to cellular senescence and organ fibrosis. Pharmacologically or genetically targeting this non-canonical cGAS-STING pathway attenuated lung and kidney fibrosis. Collectively, the findings identify an alternative innate immune pathway and its critical role in organ fibrosis, report an innate immunity-directed translation program and suggest the therapeutic potential for targeting the STING-PERK pathway in treating fibrotic diseases.


Subject(s)
Protein Serine-Threonine Kinases , Signal Transduction , Cellular Senescence , DNA/metabolism , Endoplasmic Reticulum/metabolism , Fibrosis , Humans , Immunity, Innate , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Protein Biosynthesis , Pyruvate Kinase/metabolism , Signal Transduction/physiology , eIF-2 Kinase
8.
Mol Cell ; 81(20): 4147-4164.e7, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34453890

ABSTRACT

Missense mutations of the tumor suppressor Neurofibromin 2 (NF2/Merlin/schwannomin) result in sporadic to frequent occurrences of tumorigenesis in multiple organs. However, the underlying pathogenicity of NF2-related tumorigenesis remains mostly unknown. Here we found that NF2 facilitated innate immunity by regulating YAP/TAZ-mediated TBK1 inhibition. Unexpectedly, patient-derived individual mutations in the FERM domain of NF2 (NF2m) converted NF2 into a potent suppressor of cGAS-STING signaling. Mechanistically, NF2m gained extreme associations with IRF3 and TBK1 and, upon innate nucleic acid sensing, was directly induced by the activated IRF3 to form cellular condensates, which contained the PP2A complex, to eliminate TBK1 activation. Accordingly, NF2m robustly suppressed STING-initiated antitumor immunity in cancer cell-autonomous and -nonautonomous murine models, and NF2m-IRF3 condensates were evident in human vestibular schwannomas. Our study reports phase separation-mediated quiescence of cGAS-STING signaling by a mutant tumor suppressor and reveals gain-of-function pathogenesis for NF2-related tumors by regulating antitumor immunity.


Subject(s)
Immunity, Innate , Membrane Proteins/metabolism , Mutation, Missense , Neoplasms/metabolism , Neurofibromin 2/metabolism , Nucleotidyltransferases/metabolism , Tumor Escape , Animals , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , HEK293 Cells , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Male , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Neurofibromin 2/genetics , Nucleotidyltransferases/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction
9.
PLoS Biol ; 19(2): e3001122, 2021 02.
Article in English | MEDLINE | ID: mdl-33630828

ABSTRACT

The Hippo-YAP pathway responds to diverse environmental cues to manage tissue homeostasis, organ regeneration, tumorigenesis, and immunity. However, how phosphatase(s) directly target Yes-associated protein (YAP) and determine its physiological activity are still inconclusive. Here, we utilized an unbiased phosphatome screening and identified protein phosphatase magnesium-dependent 1A (PPM1A/PP2Cα) as the bona fide and physiological YAP phosphatase. We found that PPM1A was associated with YAP/TAZ in both the cytoplasm and the nucleus to directly eliminate phospho-S127 on YAP, which conferring YAP the nuclear distribution and transcription potency. Accordingly, genetic ablation or depletion of PPM1A in cells, organoids, and mice elicited an enhanced YAP/TAZ cytoplasmic retention and resulted in the diminished cell proliferation, severe gut regeneration defects in colitis, and impeded liver regeneration upon injury. These regeneration defects in murine model were largely rescued via a genetic large tumor suppressor kinase 1 (LATS1) deficiency or the pharmacological inhibition of Hippo-YAP signaling. Therefore, we identify a physiological phosphatase of YAP/TAZ, describe its critical effects in YAP/TAZ cellular distribution, and demonstrate its physiological roles in mammalian organ regeneration.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Protein Phosphatase 2C/metabolism , Regeneration/physiology , Transcription Factors/metabolism , Animals , Cell Proliferation , Cells, Cultured , Colitis/pathology , Humans , Intestines/physiology , Liver Regeneration/physiology , Mice, Inbred C57BL , Mice, Knockout , Organoids , Protein Phosphatase 2C/genetics , Signal Transduction , YAP-Signaling Proteins
10.
Nat Commun ; 11(1): 5762, 2020 11 13.
Article in English | MEDLINE | ID: mdl-33188184

ABSTRACT

Occurrence of Colorectal cancer (CRC) is relevant with gut microbiota. However, role of IRF3, a key signaling mediator in innate immune sensing, has been barely investigated in CRC. Here, we unexpectedly found that the IRF3 deficient mice are hyper-susceptible to the development of intestinal tumor in AOM/DSS and Apcmin/+ models. Genetic ablation of IRF3 profoundly promotes the proliferation of intestinal epithelial cells via aberrantly activating Wnt signaling. Mechanically, IRF3 in resting state robustly associates with the active ß-catenin in the cytoplasm, thus preventing its nuclear translocation and cell proliferation, which can be relieved upon microbe-induced activation of IRF3. In accordance, the survival of CRC is clinically correlated with the expression level of IRF3. Therefore, our study identifies IRF3 as a negative regulator of the Wnt/ß-catenin pathway and a potential prognosis marker for Wnt-related tumorigenesis, and describes an intriguing link between gut microbiota and CRC via the IRF3-ß-catenin axis.


Subject(s)
Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Nucleus/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/prevention & control , Interferon Regulatory Factor-3/metabolism , beta Catenin/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Enterocytes/metabolism , Enterocytes/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Ki-67 Antigen/metabolism , Mice, Inbred C57BL , Protein Binding , Protein Domains , Protein Transport , Survival Analysis , Wnt Signaling Pathway , beta Catenin/chemistry
11.
Mol Cell ; 80(5): 810-827.e7, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33171123

ABSTRACT

Mitochondrial morphology shifts rapidly to manage cellular metabolism, organelle integrity, and cell fate. It remains unknown whether innate nucleic acid sensing, the central and general mechanisms of monitoring both microbial invasion and cellular damage, can reprogram and govern mitochondrial dynamics and function. Here, we unexpectedly observed that upon activation of RIG-I-like receptor (RLR)-MAVS signaling, TBK1 directly phosphorylated DRP1/DNM1L, which disabled DRP1, preventing its high-order oligomerization and mitochondrial fragmentation function. The TBK1-DRP1 axis was essential for assembly of large MAVS aggregates and healthy antiviral immunity and underlay nutrient-triggered mitochondrial dynamics and cell fate determination. Knockin (KI) strategies mimicking TBK1-DRP1 signaling produced dominant-negative phenotypes reminiscent of human DRP1 inborn mutations, while interrupting the TBK1-DRP1 connection compromised antiviral responses. Thus, our findings establish an unrecognized function of innate immunity governing both morphology and physiology of a major organelle, identify a lacking loop during innate RNA sensing, and report an elegant mechanism of shaping mitochondrial dynamics.


Subject(s)
Dynamins/metabolism , Mitochondria/physiology , Protein Serine-Threonine Kinases/metabolism , RNA/metabolism , Zebrafish/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism , Dynamins/genetics , HCT116 Cells , HEK293 Cells , Humans , Male , Mice , Mice, Transgenic , Mutation , Protein Serine-Threonine Kinases/genetics , RNA/genetics , Signal Transduction/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
12.
Nat Cell Biol ; 21(8): 1027-1040, 2019 08.
Article in English | MEDLINE | ID: mdl-31332347

ABSTRACT

Sensing cytosolic DNA through the cGAS-STING pathway constitutes a widespread innate immune mechanism to monitor cellular damage and microbial invasion. Evading this surveillance is crucial in tumorigenesis, but the process remains largely unexplored. Here, we show that the receptor tyrosine kinase HER2 (also known as ErbB-2 or Neu) potently inhibits cGAS-STING signalling and prevents cancer cells from producing cytokines, entering senescence and undergoing apoptosis. HER2, but not EGFR, associates strongly with STING and recruits AKT1 (also known as PKB) to directly phosphorylate TBK1, which prevents the TBK1-STING association and TBK1 K63-linked ubiquitination, thus attenuating STING signalling. Unexpectedly, we observed that DNA sensing robustly activates the HER2-AKT1 axis, resulting in negative feedback. Accordingly, genetic or pharmacological targeting of the HER2-AKT1 cascade augments damage-induced cellular senescence and apoptosis, and enhances STING-mediated antiviral and antitumour immunity. Thus, our findings reveal a critical function of the oncogenic pathway in innate immune regulation and unexpectedly connect HER2-AKT1 signalling to the surveillance of cellular damage and antitumour immunity.


Subject(s)
Immunity, Innate/immunology , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/metabolism , Host-Pathogen Interactions , Humans , Membrane Proteins/immunology , Phosphorylation , Proto-Oncogene Proteins c-akt/immunology , Receptor, ErbB-2/immunology , Ubiquitination/immunology
13.
Sci Robot ; 4(33)2019 Aug 28.
Article in English | MEDLINE | ID: mdl-33137788

ABSTRACT

Small-scale soft continuum robots capable of active steering and navigation in a remotely controllable manner hold great promise in diverse areas, particularly in medical applications. Existing continuum robots, however, are often limited to millimeter or centimeter scales due to miniaturization challenges inherent in conventional actuation mechanisms, such as pulling mechanical wires, inflating pneumatic or hydraulic chambers, or embedding rigid magnets for manipulation. In addition, the friction experienced by the continuum robots during navigation poses another challenge for their applications. Here, we present a submillimeter-scale, self-lubricating soft continuum robot with omnidirectional steering and navigating capabilities based on magnetic actuation, which are enabled by programming ferromagnetic domains in its soft body while growing hydrogel skin on its surface. The robot's body, composed of a homogeneous continuum of a soft polymer matrix with uniformly dispersed ferromagnetic microparticles, can be miniaturized below a few hundreds of micrometers in diameter, and the hydrogel skin reduces the friction by more than 10 times. We demonstrate the capability of navigating through complex and constrained environments, such as a tortuous cerebrovascular phantom with multiple aneurysms. We further demonstrate additional functionalities, such as steerable laser delivery through a functional core incorporated in the robot's body. Given their compact, self-contained actuation and intuitive manipulation, our ferromagnetic soft continuum robots may open avenues to minimally invasive robotic surgery for previously inaccessible lesions, thereby addressing challenges and unmet needs in healthcare.

14.
Cell Host Microbe ; 21(6): 754-768.e5, 2017 Jun 14.
Article in English | MEDLINE | ID: mdl-28618271

ABSTRACT

Cytosolic nucleic acid sensing elicits interferon production for primary antiviral defense through cascades controlled by protein ubiquitination and Ser/Thr phosphorylation. Here we show that TBK1, a core kinase of antiviral pathways, is inhibited by tyrosine phosphorylation. The Src family kinases (SFKs) Lck, Hck, and Fgr directly phosphorylate TBK1 at Tyr354/394, to prevent TBK1 dimerization and activation. Accordingly, antiviral sensing and resistance were substantially enhanced in Lck/Hck/Fgr triple knockout cells and ectopic expression of Lck/Hck/Fgr dampened the antiviral defense in cells and zebrafish. Small-molecule inhibitors of SFKs, which are conventional anti-tumor therapeutics, enhanced antiviral responses and protected zebrafish and mice from viral attack. Viral infection induced the expression of Lck/Hck/Fgr through TBK1-mediated mobilization of IRF3, thus constituting a negative feedback loop. These findings unveil the negative regulation of TBK1 via tyrosine phosphorylation and the functional integration of SFKs into innate antiviral immunity.


Subject(s)
Antiviral Agents/immunology , Immunity, Innate , Protein Serine-Threonine Kinases/metabolism , Tyrosine/metabolism , Virus Diseases/immunology , src-Family Kinases/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antiviral Agents/metabolism , Cell Line , Chlorocebus aethiops , Cytosol/immunology , Cytosol/metabolism , HEK293 Cells , Hep G2 Cells , Herpesvirus 1, Human , Humans , Interferon Regulatory Factor-3/metabolism , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-hck/metabolism , Respirovirus Infections/immunology , Rhabdoviridae Infections/immunology , Sendai virus/pathogenicity , Ubiquitination , Vero Cells , Vesiculovirus , Zebrafish/immunology , Zebrafish Proteins/metabolism , src-Family Kinases/metabolism
15.
Nat Cell Biol ; 19(4): 362-374, 2017 04.
Article in English | MEDLINE | ID: mdl-28346439

ABSTRACT

The Hippo pathway senses cellular conditions and regulates YAP/TAZ to control cellular and tissue homeostasis, while TBK1 is central for cytosolic nucleic acid sensing and antiviral defence. The correlation between cellular nutrient/physical status and host antiviral defence is interesting but not well understood. Here we find that YAP/TAZ act as natural inhibitors of TBK1 and are vital for antiviral physiology. Independent of transcriptional regulation and through the transactivation domain, YAP/TAZ associate directly with TBK1 and abolish virus-induced TBK1 activation, by preventing TBK1 Lys63-linked ubiquitylation and the binding of adaptors/substrates. Accordingly, YAP/TAZ deletion/depletion or cellular conditions inactivating YAP/TAZ through Lats1/2 kinases relieve TBK1 suppression and boost antiviral responses, whereas expression of the transcriptionally inactive YAP dampens cytosolic RNA/DNA sensing and weakens the antiviral defence in cells and zebrafish. Thus, we describe a function of YAP/TAZ and the Hippo pathway in innate immunity, by linking cellular nutrient/physical status to antiviral host defence.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cytosol/metabolism , Nucleic Acids/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Transcription Factors/metabolism , Acyltransferases , Adaptor Proteins, Signal Transducing/chemistry , Animals , Antiviral Agents/pharmacology , DNA/metabolism , Doxorubicin/pharmacology , Fluorescent Antibody Technique , Gene Expression Regulation/drug effects , HEK293 Cells , Hippo Signaling Pathway , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Lysine/metabolism , Mice, Inbred C57BL , Phosphoproteins/chemistry , Protein Binding/drug effects , Protein Domains , RNA/metabolism , Transcription, Genetic/drug effects , Transcriptional Activation/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitination/drug effects , YAP-Signaling Proteins , Zebrafish/embryology
16.
Genes Dev ; 30(9): 1086-100, 2016 05 01.
Article in English | MEDLINE | ID: mdl-27125670

ABSTRACT

Cytosolic RNA/DNA sensing elicits primary defense against viral pathogens. Interferon regulatory factor 3 (IRF3), a key signal mediator/transcriptional factor of the antiviral-sensing pathway, is indispensible for interferon production and antiviral defense. However, how the status of IRF3 activation is controlled remains elusive. Through a functional screen of the human kinome, we found that mammalian sterile 20-like kinase 1 (Mst1), but not Mst2, profoundly inhibited cytosolic nucleic acid sensing. Mst1 associated with IRF3 and directly phosphorylated IRF3 at Thr75 and Thr253. This Mst1-mediated phosphorylation abolished activated IRF3 homodimerization, its occupancy on chromatin, and subsequent IRF3-mediated transcriptional responses. In addition, Mst1 also impeded virus-induced activation of TANK-binding kinase 1 (TBK1), further attenuating IRF3 activation. As a result, Mst1 depletion or ablation enabled an enhanced antiviral response and defense in cells and mice. Therefore, the identification of Mst1 as a novel physiological negative regulator of IRF3 activation provides mechanistic insights into innate antiviral defense and potential antiviral prevention strategies.


Subject(s)
Cytosol/immunology , Immunity, Innate/genetics , Interferon Regulatory Factor-3/metabolism , Protein Serine-Threonine Kinases/metabolism , Rhabdoviridae Infections/enzymology , Rhabdoviridae Infections/immunology , Animals , Cell Line , Enzyme Activation/genetics , HEK293 Cells , Humans , Interferon Regulatory Factor-3/genetics , Mice , Mice, Inbred C57BL , Phosphorylation , Protein Binding , Serine-Threonine Kinase 3 , Vesiculovirus/immunology , Zebrafish/immunology
SELECTION OF CITATIONS
SEARCH DETAIL