Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 3533, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38670937

ABSTRACT

Oxygen is essential for aerobic organisms, but little is known about its role in antiviral immunity. Here, we report that during responses to viral infection, hypoxic conditions repress antiviral-responsive genes independently of HIF signaling. EGLN1 is identified as a key mediator of the oxygen enhancement of antiviral innate immune responses. Under sufficient oxygen conditions, EGLN1 retains its prolyl hydroxylase activity to catalyze the hydroxylation of IRF3 at proline 10. This modification enhances IRF3 phosphorylation, dimerization and nuclear translocation, leading to subsequent IRF3 activation. Furthermore, mice and zebrafish with Egln1 deletion, treatment with the EGLN inhibitor FG4592, or mice carrying an Irf3 P10A mutation are more susceptible to viral infections. These findings not only reveal a direct link between oxygen and antiviral responses, but also provide insight into the mechanisms by which oxygen regulates innate immunity.


Subject(s)
Hypoxia-Inducible Factor-Proline Dioxygenases , Immunity, Innate , Interferon Regulatory Factor-3 , Oxygen , Proline , Zebrafish , Animals , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Interferon Regulatory Factor-3/metabolism , Hydroxylation , Humans , Proline/metabolism , Mice , Oxygen/metabolism , HEK293 Cells , Phosphorylation , Mice, Knockout , Signal Transduction , Mice, Inbred C57BL
2.
Proc Natl Acad Sci U S A ; 121(17): e2314201121, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38635631

ABSTRACT

To effectively protect the host from viral infection while avoiding excessive immunopathology, the innate immune response must be tightly controlled. However, the precise regulation of antiviral innate immunity and the underlying mechanisms remain unclear. Here, we find that sirtuin3 (SIRT3) interacts with mitochondrial antiviral signaling protein (MAVS) to catalyze MAVS deacetylation at lysine residue 7 (K7), which promotes MAVS aggregation, as well as TANK-binding kinase I and IRF3 phosphorylation, resulting in increased MAVS activation and enhanced type I interferon signaling. Consistent with these findings, loss of Sirt3 in mice and zebrafish renders them more susceptible to viral infection compared to their wild-type (WT) siblings. However, Sirt3 and Sirt5 double-deficient mice exhibit the same viral susceptibility as their WT littermates, suggesting that loss of Sirt5 in Sirt3-deficient mice may counteract the increased viral susceptibility displayed in Sirt3-deficient mice. Thus, we not only demonstrate that SIRT3 positively regulates antiviral immunity in vitro and in vivo, likely via MAVS, but also uncover a previously unrecognized mechanism by which SIRT3 acts as an accelerator and SIRT5 as a brake to orchestrate antiviral innate immunity.


Subject(s)
Sirtuin 3 , Sirtuins , Virus Diseases , Animals , Mice , Adaptor Proteins, Signal Transducing/genetics , Immunity, Innate , Lysine , Sirtuin 3/genetics , Sirtuins/genetics , Zebrafish , Zebrafish Proteins
4.
Gene ; 893: 147935, 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38381506

ABSTRACT

Hypoxia, an inadequate supply of tissue oxygen tension, has been reported to induce apoptosis of spermatogenic cells and is associated with male infertility. Neddylation, a post-translational modification similar to ubiquitination, has been shown to be involved in the hypoxia stress response. However, the functions of neddylation in hypoxia-induced apoptosis of spermatogenic cells and its association with male infertility remain largely unexplored. In this study, aiming to explore the role of neddylation in male infertility, we used the specific neddylation inhibitor MLN4924 for treatment in mouse type B spermatogonia GC-2 cells. Our results showed that MLN4924 had no apparent effect on GC-2 cell apoptosis under normoxia, but significantly increased apoptotic cells under hypoxia. Transcriptomic analysis and qPCR assay confirmed that MLN4924 could suppress the expression of hypoxia target genes in GC-2 cells under hypoxia. In addition, MLN4924 could enhance the induction of intracellular and mitochondrial reactive oxygen species (ROS) under hypoxia. These results indicate that the neddylation inhibitor MLN4924 potentiates hypoxia-induced apoptosis of mouse type B spermatogonia GC-2 cells, and neddylation may play an important role in promoting spermatogenic cells to adapt to hypoxia stress.


Subject(s)
Cyclopentanes , Infertility, Male , Pyrimidines , Spermatogonia , Male , Animals , Mice , Humans , Apoptosis , Hypoxia
5.
Cell Rep ; 43(1): 113606, 2024 01 23.
Article in English | MEDLINE | ID: mdl-38127621

ABSTRACT

Activation of type I interferon (IFN-1) signaling is essential to protect host cells from viral infection. The full spectrum of IFN-I induction requires the activation of a number of cellular factors, including IκB kinase epsilon (IKKϵ). However, the regulation of IKKϵ activation in response to viral infection remains largely unknown. Here, we show that factor inhibiting hypoxia-inducible factor (HIF) (FIH), an asparaginyl hydroxylase, interacts with IKKϵ and catalyzes asparagine hydroxylation of IKKϵ at Asn-254, Asn-700, and Asn-701, resulting in the suppression of IKKϵ activation. FIH-mediated hydroxylation of IKKϵ prevents IKKϵ binding to TBK1 and TRAF3 and attenuates the cIAP1/cIAP2/TRAF2 E3 ubiquitin ligase complex-catalyzed K63-linked polyubiquitination of IKKϵ at Lys-416. In addition, Fih-deficient mice and zebrafish are more resistant to viral infection. This work uncovers a previously unrecognized role of FIH in suppressing IKKϵ activation for IFN signaling and antiviral immune responses.


Subject(s)
I-kappa B Kinase , Virus Diseases , Animals , Mice , I-kappa B Kinase/metabolism , Repressor Proteins/metabolism , Amino Acid Sequence , Hydroxylation , Zebrafish/metabolism , Immunity, Innate
6.
J Biol Chem ; 299(12): 105420, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37923141

ABSTRACT

Prolyl hydroxylase domain (PHD)-containing enzyme 3 (PHD3) belongs to the Caenorhabditis elegans gene egl-9 family of prolyl hydroxylases. PHD3 catalyzes proline hydroxylation of hypoxia-inducible factor α (HIF-α) and promotes HIF-α proteasomal degradation through coordination with the pVHL complex under normoxic conditions. However, the relationship between PHD3 and the hypoxic response is not well understood. In this study, we used quantitative real-time PCR assay and O-dianisidine staining to characterize the hypoxic response in zebrafish deficient in phd3. We found that the hypoxia-responsive genes are upregulated and the number of erythrocytes was increased in phd3-null zebrafish compared with their wild-type siblings. On the other hand, we show overexpression of phd3 suppresses HIF-transcriptional activation. In addition, we demonstrate phd3 promotes polyubiquitination of zebrafish hif-1/2α proteins, leading to their proteasomal degradation. Finally, we found that compared with wild-type zebrafish, phd3-null zebrafish are more resistant to hypoxia treatment. Therefore, we conclude phd3 has a role in hypoxia tolerance. These results highlight the importance of modulation of the hypoxia signaling pathway by phd3 in hypoxia adaptation.


Subject(s)
Hypoxia-Inducible Factor-Proline Dioxygenases , Oxygen , Procollagen-Proline Dioxygenase , Zebrafish Proteins , Zebrafish , Animals , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Procollagen-Proline Dioxygenase/genetics , Procollagen-Proline Dioxygenase/metabolism , Proline/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Gene Deletion , Oxygen/metabolism
7.
Med Sci Monit ; 29: e939858, 2023 Aug 23.
Article in English | MEDLINE | ID: mdl-37608539

ABSTRACT

BACKGROUND Patients experience severe pain in early postoperative rehabilitation after total knee arthroplasty (TKA). This study aimed to compare the effect of femoral nerve block with different concentrations of chloroprocaine on postoperative rehabilitation in patients with TKA. MATERIAL AND METHODS Ninety patients who only received unilateral TKA were randomly and equally divided into C1 (1% chloroprocaine 0.2 ml/kg), C2 (2% chloroprocaine 0.2 ml/kg), or NS (0.9% sodium chloride solution 0.2 ml/kg) groups. The patients received rehabilitation 3 times a day on days 3-6 after surgery, and femoral nerve block was performed with corresponding solution 10 min before each training session. We recorded the maximum knee flexion angles (MKFA) and maximum knee extension angles (MKEA) during active exercise on day 7 after surgery, as well as the incidence of MKFA ³100°, American knee society (AKS) scores, and postoperative rehabilitation satisfaction. Adverse effects after administration in each group were also recorded. RESULTS Compared with group NS, patients in group C1 and C2 had larger MKFA during active exercise on day 7 after TKA, and had better rehabilitation satisfaction (P<0.05). MKEA, the incidence of MKFA ≥100°, and AKS scores showed no significant differences in the 3 groups. There were more patients with decline of muscle strength in group C2 (P<0.05), and no other adverse reactions were recorded. CONCLUSIONS Chloroprocaine for femoral nerve block can be safely used in rehabilitation after TKA and to improve the knee flexion angle in the early postoperative period. Because they may have fewer adverse effects, 1% chloroprocaine 0.2 ml/kg may be preferred.


Subject(s)
Arthroplasty, Replacement, Knee , Drug-Related Side Effects and Adverse Reactions , Humans , Femoral Nerve , Procaine/therapeutic use , Knee Joint/surgery
8.
J Biol Chem ; 299(8): 105074, 2023 08.
Article in English | MEDLINE | ID: mdl-37481210

ABSTRACT

SIRT7 is a member of the sirtuin family proteins with nicotinamide adenine dinucleotide (NAD+)-dependent histone deacetylase activity, which can inhibit the activity of hypoxia-inducible factors independently of its enzymatic activity. However, the role of SIRT7 in affecting hypoxia signaling in vivo is still elusive. Here, we find that sirt7-null zebrafish are more resistant to hypoxic conditions, along with an increase of hypoxia-responsive gene expression and erythrocyte numbers, compared with their wildtype siblings. Overexpression of sirt7 suppresses the expression of hypoxia-responsive genes. Further assays indicate that sirt7 interacts with zebrafish hif-1αa, hif-1αb, hif-2αa, and hif-2αb to inhibit their transcriptional activity and mediate their protein degradation. In addition, sirt7 not only binds to the hypoxia responsive element of hypoxia-inducible gene promoters but also causes a reduction of H3K18Ac on these promoters. Sirt7 may regulate hypoxia-responsive gene expression through its enzymatic and nonenzymatic activities. This study provides novel insights into sirt7 function and sheds new light on the regulation of hypoxia signaling by sirt7.


Subject(s)
Oxygen , Sirtuins , Zebrafish Proteins , Zebrafish , Animals , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Proteolysis , Sirtuins/genetics , Sirtuins/metabolism , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Anaerobiosis , Oxygen/metabolism
9.
J Biol Chem ; 299(4): 103054, 2023 04.
Article in English | MEDLINE | ID: mdl-36822329

ABSTRACT

The deubiquitinating enzyme OTUB1 possesses canonical deubiquitinase (DUB) activity and noncanonical, catalytic-independent activity, which has been identified as an essential regulator of diverse physiological processes. Posttranslational modifications of OTUB1 affect both its DUB activity and its noncanonical activity of binding to the E2 ubiquitin-conjugation enzyme UBC13, but further investigation is needed to characterize the full inventory of modifications to OTUB1. Here, we demonstrate that SET7, a lysine monomethylase, directly interacts with OTUB1 to catalyze OTUB1 methylation at lysine 122. This modification does not affect DUB activity of OTUB1 but impairs its noncanonical activity, binding to UBC13. Moreover, we found using cell viability analysis and intracellular reactive oxygen species assay that SET7-mediated methylation of OTUB1 relieves its suppressive role on ferroptosis. Notably, the methylation-mimic mutant of OTUB1 not only loses the ability to bind to UBC13 but also relieves its suppressive role on Tert-Butyl hydroperoxide-induced cell death and Cystine starvation/Erastin-induced cellular reactive oxygen species. Collectively, our data identify a novel modification of OTUB1 that is critical for inhibiting its noncanonical activity.


Subject(s)
Deubiquitinating Enzymes , Ferroptosis , Histone-Lysine N-Methyltransferase , Ubiquitin-Conjugating Enzymes , Deubiquitinating Enzymes/metabolism , Lysine/metabolism , Protein Binding , Reactive Oxygen Species/metabolism , Ubiquitination , Humans , Histone-Lysine N-Methyltransferase/metabolism
10.
FEBS Open Bio ; 13(1): 154-163, 2023 01.
Article in English | MEDLINE | ID: mdl-36383055

ABSTRACT

Hypoxia has been reported to be an important factor leading to male infertility, and it has been reported that hypoxia can induce the apoptosis of mouse spermatogenic cells. Sirtuin 3 (SIRT3) has been reported to promote the degradation of hypoxia-inducible factor 1α (HIF-1α), and thus, we hypothesized that SIRT3 may influence hypoxia-induced apoptosis of spermatogonia. In this study, we overexpressed or inhibited SIRT3 in mouse type B spermatogonia GC-2 cells and then subjected the cells to hypoxia or normoxia, before examining hypoxia-responsive gene expression and cell viability. We report that SIRT3 stabilizes hypoxia-inducible factor 1α (HIF-1α) and activates its downstream target gene expression in GC-2 cells. We also show that the SIRT3 inhibitor 3-TYP suppresses HIF-1α target gene expression and alleviates hypoxia-induced apoptosis of GC-2 cells. Our study reveals the critical role and underlying mechanisms of SIRT3 in hypoxia-induced apoptosis of mouse type B spermatogonia GC-2 cells.


Subject(s)
Sirtuin 3 , Mice , Male , Animals , Sirtuin 3/genetics , Sirtuin 3/metabolism , Spermatogonia/metabolism , Apoptosis , Hypoxia/metabolism , Cell Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
11.
J Biol Chem ; 298(12): 102633, 2022 12.
Article in English | MEDLINE | ID: mdl-36273580

ABSTRACT

Hypoxia-inducible factor (HIF)1α, a main transcriptional regulator of the cellular response to hypoxia, also plays important roles in oxygen homeostasis of aerobic organisms, which is regulated by multiple mechanisms. However, the full cellular response to hypoxia has not been elucidated. In this study, we found that expression of SMYD3, a methyltransferase, augments hypoxia signaling independent of its enzymatic activity. We demonstrated SMYD3 binds to and stabilizes HIF1α via co-immunoprecipitation and Western blot assays, leading to the enhancement of HIF1α transcriptional activity under hypoxia conditions. In addition, the stabilization of HIF1α by SMYD3 is independent of HIF1α hydroxylation by prolyl hydroxylases and the intactness of the von Hippel-Lindau ubiquitin ligase complex. Furthermore, we showed SMYD3 induces reactive oxygen species accumulation and promotes hypoxia-induced cell apoptosis. Consistent with these results, we found smyd3-null zebrafish exhibit higher hypoxia tolerance compared to their wildtype siblings. Together, these findings define a novel role of SMYD3 in affecting hypoxia signaling and demonstrate that SMYD3-mediated HIF1α stabilization augments hypoxia signaling, leading to the impairment of hypoxia tolerance.


Subject(s)
Histone-Lysine N-Methyltransferase , Hypoxia , Methyltransferases , Zebrafish Proteins , Animals , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Methyltransferases/metabolism , Signal Transduction , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Zebrafish/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
12.
J Immunol ; 209(6): 1165-1172, 2022 09 15.
Article in English | MEDLINE | ID: mdl-36002231

ABSTRACT

The signaling adaptor MAVS is a critical determinant in retinoic acid-inducible gene 1-like receptor signaling, and its activation is tightly controlled by multiple mechanisms in response to viral infection, including phosphorylation and ubiquitination. In this article, we demonstrate that zebrafish sirt5, one of the sirtuin family proteins, negatively regulates mavs-mediated antiviral innate immunity. Sirt5 is induced by spring viremia of carp virus (SVCV) infection and binds to mavs, resulting in attenuating phosphorylation and ubiquitination of mavs. Disruption of sirt5 in zebrafish promotes survival ratio after challenge with SVCV. Consistently, the antiviral responsive genes are enhanced, and the replication of SVCV is diminished in sirt5-dificient zebrafish. Therefore, we reveal a function of zebrafish sirt5 in the negative regulation of antiviral innate immunity by targeting mavs.


Subject(s)
Sirtuins , Zebrafish , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antiviral Agents , Immunity, Innate , Phosphorylation , Rhabdoviridae , Sirtuins/metabolism , Tretinoin/metabolism , Ubiquitination , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
13.
Cell Rep ; 39(10): 110920, 2022 06 07.
Article in English | MEDLINE | ID: mdl-35675783

ABSTRACT

Retinoic acid-inducible-I (RIG-I), melanoma differentiation-associated gene 5 (MDA5), and cyclic GMP-AMP synthase (cGAS) genes encode essential cytosolic receptors mediating antiviral immunity against viruses. Here, we show that OTUD3 has opposing role in response to RNA and DNA virus infection by removing distinct types of RIG-I/MDA5 and cGAS polyubiquitination. OTUD3 binds to RIG-I and MDA5 and removes K63-linked ubiquitination. This serves to reduce the binding of RIG-I and MDA5 to viral RNA and the downstream adaptor MAVS, leading to the suppression of the RNA virus-triggered innate antiviral responses. Meanwhile, OTUD3 associates with cGAS and targets at Lys279 to deubiquitinate K48-linked ubiquitination, resulting in the enhancement of cGAS protein stability and DNA-binding ability. As a result, Otud3-deficient mice and zebrafish are more resistant to RNA virus infection but are more susceptible to DNA virus infection. These findings demonstrate that OTUD3 limits RNA virus-triggered innate immunity but promotes DNA virus-triggered innate immunity.


Subject(s)
DNA Virus Infections , Immunity, Innate , RNA Virus Infections , Ubiquitin-Specific Proteases , Animals , DEAD Box Protein 58/metabolism , DNA Virus Infections/immunology , DNA Viruses , Deubiquitinating Enzymes , Interferon-Induced Helicase, IFIH1/metabolism , Mice , Nucleotidyltransferases , RNA Virus Infections/immunology , RNA Viruses , RNA, Viral/metabolism , Ubiquitin-Specific Proteases/metabolism , Zebrafish/metabolism
14.
Cell Death Dis ; 13(6): 560, 2022 06 22.
Article in English | MEDLINE | ID: mdl-35732631

ABSTRACT

As a main regulator of cellular responses to hypoxia, the protein stability of hypoxia-inducible factor (HIF)-1α is strictly controlled by oxygen tension dependent of PHDs-catalyzed protein hydroxylation and pVHL complex-mediated proteasomal degradation. Whether HIF-1α protein stability as well as its activity can be further regulated under hypoxia is not well understood. In this study, we found that OTUB1 augments hypoxia signaling independent of PHDs/VHL and FIH. OTUB1 binds to HIF-1α and depletion of OTUB1 reduces endogenous HIF-1α protein under hypoxia. In addition, OTUB1 inhibits K48-linked polyubiquitination of HIF-1α via its non-canonical inhibition of ubiquitination activity. Furthermore, OTUB1 promotes hypoxia-induced glycolytic reprogramming for cellular metabolic adaptation. These findings define a novel regulation of HIF-1α under hypoxia and demonstrate that OTUB1-mediated HIF-1α stabilization positively regulates HIF-1α transcriptional activity and benefits cellular hypoxia adaptation.


Subject(s)
Cell Hypoxia , Deubiquitinating Enzymes , Hypoxia-Inducible Factor 1, alpha Subunit , Signal Transduction , Cell Hypoxia/physiology , Deubiquitinating Enzymes/genetics , Deubiquitinating Enzymes/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Ubiquitination
15.
J Biol Chem ; 298(6): 101961, 2022 06.
Article in English | MEDLINE | ID: mdl-35452683

ABSTRACT

Egg-laying defective nine 1 (EGLN1) functions as an oxygen sensor to catalyze prolyl hydroxylation of the transcription factor hypoxia-inducible factor-1 α under normoxia conditions, leading to its proteasomal degradation. Thus, EGLN1 plays a central role in the hypoxia-inducible factor-mediated hypoxia signaling pathway; however, the posttranslational modifications that control EGLN1 function remain largely unknown. Here, we identified that a lysine monomethylase, SET7, catalyzes EGLN1 methylation on lysine 297, resulting in the repression of EGLN1 activity in catalyzing prolyl hydroxylation of hypoxia-inducible factor-1 α. Notably, we demonstrate that the methylation mimic mutant of EGLN1 loses the capability to suppress the hypoxia signaling pathway, leading to the enhancement of cell proliferation and the oxygen consumption rate. Collectively, our data identify a novel modification of EGLN1 that is critical for inhibiting its enzymatic activity and which may benefit cellular adaptation to conditions of hypoxia.


Subject(s)
Histone-Lysine N-Methyltransferase , Hypoxia-Inducible Factor 1, alpha Subunit , Hypoxia-Inducible Factor-Proline Dioxygenases , Lysine , Animals , Catalysis , Humans , Hydroxylation , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Lysine/metabolism , Methylation , Oxygen/metabolism , Protein Processing, Post-Translational
16.
Cell Death Differ ; 29(4): 722-736, 2022 04.
Article in English | MEDLINE | ID: mdl-34642466

ABSTRACT

p53 is a classic tumor suppressor that functions in maintaining genome stability by inducing either cell arrest for damage repair or cell apoptosis to eliminate damaged cells in response to different types of stress. Posttranslational modifications (PTMs) of p53 are thought to be the most effective way for modulating of p53 activation. Here, we show that SIRT5 interacts with p53 and suppresses its transcriptional activity. Using mass spectrometric analysis, we identify a previously unknown PTM of p53, namely, succinylation of p53 at Lysine 120 (K120). SIRT5 mediates desuccinylation of p53 at K120, resulting in the suppression of p53 activation. Moreover, using double knockout mice (p53-/-Sirt5-/-), we validate that the suppression of p53 target gene expression and cell apoptosis upon DNA damage is dependent on cellular p53. Our study identifies a novel PTM of p53 that regulates its activation as well as reveals a new target of SIRT5 acting as a desuccinylase.


Subject(s)
Lysine , Protein Processing, Post-Translational , Sirtuins , Tumor Suppressor Protein p53 , Animals , DNA Damage , Lysine/metabolism , Mice , Mice, Knockout , Sirtuins/genetics , Sirtuins/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
17.
Mol Cell ; 81(15): 3171-3186.e8, 2021 08 05.
Article in English | MEDLINE | ID: mdl-34171297

ABSTRACT

Accurate control of innate immune responses is required to eliminate invading pathogens and simultaneously avoid autoinflammation and autoimmune diseases. Here, we demonstrate that arginine monomethylation precisely regulates the mitochondrial antiviral-signaling protein (MAVS)-mediated antiviral response. Protein arginine methyltransferase 7 (PRMT7) forms aggregates to catalyze MAVS monomethylation at arginine residue 52 (R52), attenuating its binding to TRIM31 and RIG-I, which leads to the suppression of MAVS aggregation and subsequent activation. Upon virus infection, aggregated PRMT7 is disabled in a timely manner due to automethylation at arginine residue 32 (R32), and SMURF1 is recruited to PRMT7 by MAVS to induce proteasomal degradation of PRMT7, resulting in the relief of PRMT7 suppression of MAVS activation. Therefore, we not only reveal that arginine monomethylation by PRMT7 negatively regulates MAVS-mediated antiviral signaling in vitro and in vivo but also uncover a mechanism by which PRMT7 is tightly controlled to ensure the timely activation of antiviral defense.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Arginine/metabolism , Host-Pathogen Interactions/physiology , Immunity, Innate/physiology , Protein-Arginine N-Methyltransferases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Animals , DEAD Box Protein 58/metabolism , Fibroblasts/virology , HEK293 Cells , Herpes Simplex/immunology , Herpes Simplex/metabolism , Herpes Simplex/virology , Humans , Methylation , Mice , Mice, Knockout , Polyunsaturated Alkamides , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/immunology , Receptors, Immunologic/metabolism , Respirovirus Infections/immunology , Respirovirus Infections/metabolism , Respirovirus Infections/virology , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
18.
EMBO J ; 39(11): e103285, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32301534

ABSTRACT

RLR-mediated type I IFN production plays a pivotal role in innate antiviral immune responses, where the signaling adaptor MAVS is a critical determinant. Here, we show that MAVS is a physiological substrate of SIRT5. Moreover, MAVS is succinylated upon viral challenge, and SIRT5 catalyzes desuccinylation of MAVS. Mass spectrometric analysis indicated that Lysine 7 of MAVS is succinylated. SIRT5-catalyzed desuccinylation of MAVS at Lysine 7 diminishes the formation of MAVS aggregation after viral infection, resulting in the inhibition of MAVS activation and leading to the impairment of type I IFN production and antiviral gene expression. However, the enzyme-deficient mutant of SIRT5 (SIRT5-H158Y) loses its suppressive role on MAVS activation. Furthermore, we show that Sirt5-deficient mice are resistant to viral infection. Our study reveals the critical role of SIRT5 in limiting RLR signaling through desuccinylating MAVS.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Protein Aggregates , Sirtuins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Substitution , Animals , Gene Expression Regulation , HCT116 Cells , HEK293 Cells , Humans , Interferon Type I/biosynthesis , Interferon Type I/genetics , Mice , Mice, Knockout , Mutation, Missense , Sirtuins/genetics
19.
Zhonghua Yi Xue Za Zhi ; 95(38): 3129-32, 2015 Oct 13.
Article in Chinese | MEDLINE | ID: mdl-26814104

ABSTRACT

OBJECTIVE: To explore the feasibility of different doses of dexmedetomidine for quiet extubation during anesthesia recovery in hypertensive patients monitored with Narcotrend. METHODS: A total of 120 hypertensive patients scheduled for thyroid surgery from August 2012 to June 2014 in the Second Affiliated Hospital of Wenzhou Medical University, were randomly divided into 6 groups (n=20 each). Dexmedetomidine 0.4 (group M1), 0.6 (group M2), 0.8 (group M3) or 1.0 (group M4) µg·kg(-1)·h(-1), remifentanil 0.1 µg·kg(-1)·min(-1) (group R) and normal saline (group S) were infused for half an hour before the end of surgery, and extubation was carried out when Narcotrend index (NI) values were ≥80 in each group. Data of heart rate (HR), systolic blood pressure (SBP) and minimal alveolar concentration (MAC) of sevoflurane were observed and recorded at the time of baseline (T0), half an hour (T1) and 15 min (T2) before the end of surgery, stopping sevoflurane (T3), before extubation (T4), 1 min (T5), 5 min (T6) and 10 min (T7) after extubation. Extubation time, recovery time and related adverse reactions were also recorded. RESULTS: Compared with T0, SBP and HR at T4 to T7 in four M groups were significantly lower (all P<0.05). SBP and HR at T6, T7 in group R were significantly lower than at T0 (all P<0.05). SBP at T6, T7 and HR at T4 to T7 in group R were significantly lower than that of group S (all P<0.05). SBP and HR at T4 to T7 in four M groups were significantly lower than that of group S and group R (all P<0.05). The values of MAC of sevoflurane at T2 and T3 in group R and M2-4 were significantly lower than at T1 and that of group S (all P<0.05). Recovery time in group M3 [(19.1±2.8) min] and group M4 [(20.6±4.1) min] were significantly longer compared with other four groups (all P<0.05). The percentage of cough grade at level I and II in each M group and group R during extubation was significantly higher than that in group S (85%, 85%, 90%, 95%, 80% vs 45%, all P<0.05). CONCLUSIONS: Dexmedetomidine could be safely used in hypertensive patients monitored with Narcotrend for quiet extubation during anesthesia recovery, but larger doses of dexmedetomidine may prolong the recovery time.


Subject(s)
Airway Extubation , Anesthesia Recovery Period , Hypertension , Blood Pressure , Dexmedetomidine , Heart Rate , Humans , Methyl Ethers , Piperidines , Remifentanil , Sevoflurane
SELECTION OF CITATIONS
SEARCH DETAIL
...