Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Cell Biochem Biophys ; 2024 May 29.
Article in English | MEDLINE | ID: mdl-38809352

ABSTRACT

Temozolomide (TMZ) stands as the primary chemotherapeutic drug utilized in clinical glioma treatment, particularly for high-grade glioblastoma (GBM). However, the emergence of TMZ resistance in GBM poses a significant hurdle to its clinical efficacy. Our objective was to elucidate the role of deubiquitinating enzymes (DUBs) in GBM cell resistance to TMZ. We employed the broad-spectrum DUBs inhibitor G5 to investigate the function of DUBs in TMZ cytotoxicity against GBM cells. Eighty-two GBM cell lines with specified DUBs knockout were generated and subjected to CCK-8 assays to assess cell proliferation and TMZ resistance. Furthermore, the association between DUBs and TMZ resistance in GBM cells, along with the modulation of autophagic flux, was examined. The pan-DUBs inhibitor G5 demonstrated the ability to induce cell death and enhance TMZ toxicity in GBM cells. Subsequently, we identified potential DUBs involved in regulating GBM cell proliferation and TMZ resistance. The impact of DUBs knockout on TMZ cytotoxicity was found to be associated with their regulation of TMZ-induced autophagy. In summary, our study provides primary insights into the role of DUBs in GBM cell proliferation and TMZ resistance, and contributes to a deeper understanding of the complex function of DUBs genes underlying TMZ resistance in GBM cells.

2.
Biochem Biophys Res Commun ; 714: 149973, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38657444

ABSTRACT

Acute respiratory distress syndrome (ARDS) is characterized by acute diffuse inflammatory lung injury with a high mortality rate. Mesenchymal stromal cells (MSC) are pluripotent adult cells that can be extracted from a variety of tissues, including the lung. Lung-resident MSC (LR-MSC) located around vascular vessels and act as important regulators of lung homeostasis, regulating the balance between lung injury and repair processes. LR-MSC support the integrity of lung tissue by modulating immune responses and releasing trophic factors. Studies have reported that the STING pathway is involved in the progression of lung injury inflammation, but the specific mechanism is unclear. In this study, we found that STING deficiency could ameliorate lipopolysaccharides (LPS)-induced acute lung injury, STING knockout (STING KO) LR-MSC had an enhanced treatment effect on acute lung injury. STING depletion protected LR-MSC from LPS-induced apoptosis. RNA-sequencing and Western blot results showed that STING KO LR-MSC expressed higher levels of MSC immunoregulatory molecules, such as Igfbp4, Icam1, Hgf and Cox2, than WT LR-MSC. This study highlights that LR-MSC have a therapeutic role in acute lung injury, and we demonstrate that STING deficiency can enhance the immunomodulatory function of LR-MSC in controlling lung inflammation. Thus, STING can be used as an intervention target to enhance the therapeutic effect of MSC.


Subject(s)
Acute Lung Injury , Lipopolysaccharides , Lung , Membrane Proteins , Mesenchymal Stem Cells , Mice, Inbred C57BL , Animals , Lipopolysaccharides/toxicity , Mesenchymal Stem Cells/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/deficiency , Lung/pathology , Lung/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/pathology , Acute Lung Injury/therapy , Acute Lung Injury/metabolism , Mice , Mice, Knockout , Apoptosis , Male
3.
Genes Dis ; 10(4): 1596-1612, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37397525

ABSTRACT

Non-alcoholic steatohepatitis (NASH), a progressive form of non-alcoholic fatty liver disease (NAFLD), is characterised by chronic liver inflammation, which can further progress into complications such as liver cirrhosis and NASH-associated hepatocellular carcinoma (HCC) and therefore has become a growing health problem worldwide. The type I interferon (IFN) signaling pathway plays a pivotal role in chronic inflammation; however, the molecular mechanisms underlying NAFLD/NASH from the perspective of innate immune response has not yet been fully explored. In this study, we elucidated the mechanisms of how innate immune response modulates NAFLD/NASH pathogenesis, and demonstrated that hepatocyte nuclear factor-1alpha (HNF1A) was suppressed and the type I IFN production pathway was activated in liver tissues of patients with NAFLD/NASH. Further experiments suggested that HNF1A negatively regulates the TBK1-IRF3 signaling pathway by promoting autophagic degradation of phosphorylated-TBK1, which constrains IFN production, thereby inhibiting the activation of type I IFN signaling. Mechanistically, HNF1A interacts with the phagophore membrane protein LC3 through its LIR-docking sites, and mutations of LIRs (LIR2, LIR3, LIR4, and LIRs) block the HNF1A-LC3 interaction. In addition, HNF1A was identified not only as a novel autophagic cargo receptor but also to specifically induce K33-linked ubiquitin chains on TBK1 at Lys670, thereby resulting in autophagic degradation of TBK1. Collectively, our study illustrates the crucial function of the HNF1A-TBK1 signaling axis in NAFLD/NASH pathogenesis via cross-talk between autophagy and innate immunity.

5.
Biochem Biophys Res Commun ; 652: 88-94, 2023 04 16.
Article in English | MEDLINE | ID: mdl-36841099

ABSTRACT

Acetaminophen (APAP) overdose is the most common cause for acute liver failure (ALF) in the developed countries, with limited treatment options. Piezo1 is a mechanosensitive cation channel. We found that APAP caused upregulation of Piezo1 in both an APAP-induced acute liver injury (ALI) animal model and a mouse hepatocyte cell line AML12. Activation of Piezo1 by its activator Yoda1 reduced APAP-induced hepatotoxicity and ROS level. Mechanistically, activation of Piezo1 led to accumulation of the antioxidant regulator Nrf2 and upregulation of its target genes Nqo1 and Gsta1, while knockdown of Piezo1 downregulated this pathway. Finally, injection of Yoda1 decreased serum AST and ALT levels, reduced cell death and rescued liver injury in the APAP-induced ALI mouse model. Our findings suggested a previously undiscovered protective role of Piezo1 in APAP-induced ALI, which might shed light on a new therapeutic target for this disease.


Subject(s)
Acetaminophen , Chemical and Drug Induced Liver Injury , Animals , Mice , Reactive Oxygen Species/metabolism , NF-E2-Related Factor 2/metabolism , Liver/metabolism , Hepatocytes/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Oxidative Stress , Mice, Inbred C57BL , Ion Channels/metabolism
6.
Autophagy ; 19(3): 1026-1027, 2023 03.
Article in English | MEDLINE | ID: mdl-35944095

ABSTRACT

The selective macroautophagy/autophagy pathway is an important pathway of protein degradation, regulating signal transduction pathways via selective degradation of certain signaling complexes. TBK1 functions as a key protein in innate immunity or metabolic-associated fatty liver disease (MAFLD); however, the degradation of TBK1 has not been fully investigated. Recently, we have found that HNF1A functions as a novel cargo receptor to bridge TBK1 and MAP1LC3/LC3, hence promoting the degradation of TBK1 and regulating antiviral innate immunity and MAFLD.


Subject(s)
Autophagy , Protein Serine-Threonine Kinases , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Immunity, Innate , Phosphorylation
7.
Org Lett ; 24(31): 5693-5697, 2022 Aug 12.
Article in English | MEDLINE | ID: mdl-35921617

ABSTRACT

A Lewis-base-catalyzed three-component electrophilic thiofunctionalization of cyclopropene with phenol is developed to furnish various trisubstituted chromanes in high trans-diasteroselectivity. This metal-free protocol is easy to scale-up, offers a unique 2,2,3-substitution pattern, and delivers chromanes with diversified core substitution patterns. The unprecedented tolerance of strong electron-withdrawing substituents at the phenol renders the protocol indispensable to access the otherwise inaccessible chromane chemical space that is important for medicinal chemistry campaigns.

8.
Biochem Biophys Res Commun ; 623: 181-188, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35921710

ABSTRACT

Type I interferon pathway is a crucial component of innate immune signaling upon pathogen infection or endogenous instability. An imbalance of type I interferon can lead to many diseases, such as autoimmune diseases and inflammatory diseases. Meanwhile, the side effects of clinical drugs on type I interferon signaling may result in impaired outcomes in clinical treatment, especially in cancer immunotherapy which is associated with type I interferon signaling. Here, we found that sorafenib, an FDA-approved drug for HCC chemotherapy, suppresses both DNA- and RNA-sensing mediated type I interferon pathway. Mechanistically, sorafenib treatment induces the autophagic degradation of MAVS, cGAS, TBK1, and IRF3, and attenuates the signaling transduction. In addition, sorafenib also inhibits the recruiting of STING or MAVS with TBK1 and IRF3. This work reveals the negative role of sorafenib in the regulation of type I interferon pathway. Sorafenib treatment is not only a potential drug for autoimmune disease and inflammation diseases, but also needs to be noticed in HCC chemotherapy.


Subject(s)
Carcinoma, Hepatocellular , Interferon Type I , Liver Neoplasms , Humans , Immunity, Innate , Interferon Regulatory Factor-3/metabolism , Interferon Type I/metabolism , Nucleotidyltransferases/metabolism , Protein Serine-Threonine Kinases , Sorafenib/pharmacology
9.
J Immunother Cancer ; 10(8)2022 08.
Article in English | MEDLINE | ID: mdl-36002188

ABSTRACT

BACKGROUND: Emerging evidence indicates that the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) axis plays a pivotal role in intrinsic antitumor immunity. Previous studies demonstrate that the conventional chemotherapy agent, teniposide, effectively promotes the therapeutic efficacy of programmed cell death protein-1 antibody (PD-1 Ab) through robust cGAS-STING activation. Unfortunately, the cGAS expression of tumor cells is reported to be severely suppressed by the hypoxic status in solid tumor. Clinically, enhancing chemotherapy-induced, DNA-activated tumor STING signaling by alleviating tumor hypoxia might be one possible direction for improving the currently poor response rates of patients with hepatocellular carcinoma (HCC) to PD-1 Ab. METHODS: Teniposide was first screened out from several chemotherapy drugs according to their potency in inducing cGAS-STING signaling in human HCC cells. Teniposide-treated HCC cells were then cultured under hypoxia, normoxia or reoxygenation condition to detect change in cGAS-STING signaling. Next, oxaliplatin/teniposide chemotherapy alone or combined with hyperbaric oxygen (HBO) therapy was administered on liver orthotopic mouse tumor models, after which the tumor microenvironment (TME) was surveyed. Lastly, teniposide alone or combined with HBO was performed on multiple mouse tumor models and the subsequent anti-PD-1 therapeutic responses were observed. RESULTS: Compared with the first-line oxaliplatin chemotherapy, teniposide chemotherapy induced stronger cGAS-STING signaling in human HCC cells. Teniposide-induced cGAS-STING activation was significantly inhibited by hypoxia inducible factor 1α in an oxygen-deficient environment in vitro and the inhibition was rapidly removed via effective reoxygenation. HBO remarkably enhanced the cGAS-STING-dependent tumor type Ⅰ interferon and nuclear factor kappa-B signaling induced by teniposide in vivo, both of which contributed to the activation of dendritic cells and subsequent cytotoxic T cells. Combined HBO with teniposide chemotherapy improved the therapeutic effect of PD-1 Ab in multiple tumor models. CONCLUSIONS: By combination of two therapies approved by the Food and Drug Administration, we safely stimulated an immunogenic, T cell-inflamed HCC TME, leading to further sensitization of tumors to anti-PD-1 immunotherapy. These findings might enrich therapeutic strategies for advanced HCC andwe can attempt to improve the response rates of patients with HCC to PD-1 Ab by enhancing DNA-activated STING signaling through effective tumor reoxygenation.


Subject(s)
Carcinoma, Hepatocellular , Hyperbaric Oxygenation , Liver Neoplasms , Animals , Antibodies , Carcinoma, Hepatocellular/drug therapy , Humans , Hypoxia , Liver Neoplasms/drug therapy , Membrane Proteins , Mice , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Oxaliplatin , Oxygen , Teniposide , Tumor Microenvironment , United States
10.
Autophagy ; 18(4): 860-876, 2022 04.
Article in English | MEDLINE | ID: mdl-34382907

ABSTRACT

Lipid accumulation often leads to lipotoxic injuries to hepatocytes, which can cause nonalcoholic steatohepatitis. The association of inflammation with lipid accumulation in liver tissue has been studied for decades; however, key mechanisms have been identified only recently. In particular, it is still unknown how hepatic inflammation regulates lipid metabolism in hepatocytes. Herein, we found that PA treatment or direct stimulation of STING1 promoted, whereas STING1 deficiency impaired, MTORC1 activation, suggesting that STING1 is involved in PA-induced MTORC1 activation. Mechanistic studies revealed that STING1 interacted with several components of the MTORC1 complex and played an important role in the complex formation of MTORC1 under PA treatment. The involvement of STING1 in MTORC1 activation was dependent on SQSTM1, a key regulator of the MTORC1 pathway. In SQSTM1-deficient cells, the interaction of STING1 with the components of MTORC1 was weak. Furthermore, the impaired activity of MTORC1 via rapamycin treatment or STING1 deficiency decreased the numbers of LDs in cells. PA treatment inhibited lipophagy, which was not observed in STING1-deficient cells or rapamycin-treated cells. Restoration of MTORC1 activity via treatment with amino acids blocked lipophagy and LDs degradation. Finally, increased MTORC1 activation concomitant with STING1 activation was observed in liver tissues of nonalcoholic fatty liver disease patients, which provided clinical evidence for the involvement of STING1 in MTORC1 activation. In summary, we identified a novel regulatory loop of STING1-MTORC1 and explain how hepatic inflammation regulates lipid accumulation. Our findings may facilitate the development of new strategies for clinical treatment of hepatic steatosis.Abbreviations: AA: amino acid; ACTB: actin beta; cGAMP: cyclic GMP-AMP; CGAS: cyclic GMP-AMP synthase; DEPTOR: DEP domain containing MTOR interacting protein; EIF4EBP1: eukaryotic translation initiation factor 4E binding protein 1; FFAs: free fatty acids; GFP: green fluorescent protein; HFD: high-fat diet; HT-DNA: herring testis DNA; IL1B: interleukin 1 beta; LAMP1: lysosomal associated membrane protein 1; LDs: lipid droplets; MAP1LC3: microtubule associated protein 1 light chain 3; MAP1LC3B: microtubule associated protein 1 light chain 3 beta; MEFs: mouse embryonic fibroblasts; MLST8: MTOR associated protein, LST8 homolog; MT-ND1: mitochondrially encoded NADH: ubiquinone oxidoreductase core subunit 1; mtDNA: mitochondrial DNA; MTOR: mechanistic target of rapamycin kinase; MTORC1: MTOR complex 1; NAFL: nonalcoholic fatty liver; NAFLD: nonalcoholic fatty liver disease; NASH: nonalcoholic steatohepatitis; NPCs: non-parenchymal cells; PA: palmitic acid; PLIN2: perilipin 2; RD: regular diet; RELA: RELA proto-oncogene, NF-kB subunit; RPS6: ribosomal protein S6; RPS6KB1: ribosomal protein S6 kinase B1; RPTOR: regulatory associated protein of MTOR complex 1; RRAGA: Ras related GTP binding A; RRAGC: Ras related GTP binding C; SQSTM1: sequestosome 1; STING1: stimulator of interferon response cGAMP interactor 1; TBK1: TANK binding kinase 1; TGs: triglycerides; TREX1: three prime repair exonuclease 1.


Subject(s)
Autophagy , Non-alcoholic Fatty Liver Disease , Animals , Autophagy/physiology , Fibroblasts/metabolism , Guanosine Triphosphate , Humans , Inflammation , Intracellular Signaling Peptides and Proteins/metabolism , Lipids , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Microtubule-Associated Proteins/metabolism , Sequestosome-1 Protein/metabolism , Sirolimus
11.
Front Oncol ; 11: 711448, 2021.
Article in English | MEDLINE | ID: mdl-34888230

ABSTRACT

Accumulating evidence shows a close association between various types of bile acids (BAs) and hepatocellular carcinoma (HCC), and they have been revealed to affect tumor immune response and progression mainly by regulating Farnesoid X receptor (FXR). Nevertheless, the roles of Norcholic acid(NorCA) in HCC progression remain unknown yet. In this study, herein we demonstrate that NorCA can promote HCC cell proliferation, migration and invasion through negatively regulating FXR. Additionally, NorCA can increase PD-L1 level on the surfaces of HCC cells and their exosomes, and NorCA-induced exosomes dramatically dampen the function of CD4+T cells, thereby inducing an immunosuppressive microenvironment. Meanwhile, a negative correlation between PD-L1 and FXR expression in human HCC specimens was identified, and HCC patients with FXRlowPD-L1high expression exhibit a rather dismal survival outcome. Importantly, FXR agonist (GW4064) can synergize with anti-PD-1 antibody (Ab) to inhibit HCC growth in tumor-bearing models. Taken together, NorCA can promote HCC progression and immune invasion by inhibiting FXR signaling, implying a superiority of the combination of FXR agonist and anti-PD-1 Ab to the monotherapy of immune checkpoint inhibitor in combating HCC. However, more well-designed animal experiments and clinical trials are warranted to further confirm our findings in future due to the limitations in our study.

12.
Cell Death Discov ; 7(1): 374, 2021 Dec 04.
Article in English | MEDLINE | ID: mdl-34864819

ABSTRACT

Berberine (BBR) has been explored as a potential anti-liver fibrosis agent, but the underlying mechanisms are unknown. In the current study, we aimed to investigate the molecular mechanisms underlying the effect of BBR against liver fibrogenesis in thioacetamide (TAA) and carbon tetrachloride (CCl4) induced mouse liver fibrosis. In addition to i.p. injection with TAA or CCl4, mice in the treatment group received BBR intragastrically. Concurrently, combined with TAA and BBR treatment, mice in the inhibitor group were injected i.p. with ferrostatin-1 (Fer-1). Hepatic stellate cells (HSCs) were also used in the study. Our results showed that BBR obviously alleviated mouse liver fibrosis and restored mouse liver function; however, the pharmacological effects of BBR against liver fibrosis were significantly diminished by Fer-1 treatment. Mechanically, BBR impaired the autophagy-lysosome pathway (ALP) and increased cell reactive oxygen species (ROS) production in HSCs. ROS accelerated the breakdown of the iron-storage protein ferritin and sped up iron release from ferritin, which resulted in redox-active iron accumulation in HSCs. Lipid peroxidation and glutathione (GSH) depletion triggered by the Fenton reaction promoted ferroptosis and attenuated liver fibrosis. Furthermore, impaired autophagy enhanced BBR-mediated ferritin proteolysis to increase cellular ferrous overload via the ubiquitin-proteasome pathway (UPS) in HSCs and triggered HSC ferroptosis. Collectively, BBR alleviated liver fibrosis by inducing ferrous redox to activate ROS-mediated HSC ferroptosis. Our findings may be exploited clinically to provide a potential novel therapeutic strategy for liver fibrosis.

13.
Mol Immunol ; 129: 45-52, 2021 01.
Article in English | MEDLINE | ID: mdl-33278678

ABSTRACT

Type I interferons (IFNs) play a central role in host defense against viral infection. Multiple posttranslational modifications including ubiquitination and deubiquitination regulate the function of diverse molecules in type I IFN signaling. Many ubiquitin ligase enzymes, such as those of the TRAF and TRIM families, have been shown to participate in the production of type I IFNs and inflammatory cytokines. However, the function of deubiquitinating enzymes (DUBs), a protein family that counteracts the action of protein ubiquitination, on the regulation of antiviral immune responses is not well understood. In this study, we used the broad-spectrum DUB inhibitor G5 to reveal their function in antiviral signaling, and then systematically analyzed mRNA expression of the DUB genes upon poly (I:C) treatment in THP-1 cells. Based on this analysis, we cloned some DUB genes whose expression changed and determined their function in antiviral signaling. Taken together, we present a comprehensive DUB gene expression analysis in THP-1 cells, and suggest the involvement of this family of proteins in the regulation of host antiviral activities.


Subject(s)
Antiviral Agents/pharmacology , Deubiquitinating Enzymes/genetics , Gene Expression/drug effects , Gene Expression/genetics , Poly I-C/pharmacology , Signal Transduction/genetics , Cell Line , Cytokines/genetics , HEK293 Cells , Humans , Immunity/drug effects , Immunity/genetics , Interferon Type I/genetics , Pyrans/pharmacology , RNA, Messenger/genetics , Signal Transduction/drug effects , Sulfhydryl Compounds/pharmacology , THP-1 Cells , Ubiquitin/genetics , Ubiquitination/drug effects , Ubiquitination/genetics
14.
Chinese Journal of School Health ; (12): 742-745, 2021.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-877142

ABSTRACT

Objective@#To demonstrate the effect of structured and autonomous sports activities on children s basic motor skills in a preset outdoor field for a certain time and frequency, and to provide a reference for improving children s development of basic motor skills.@*Methods@#Selected 141 typical developing children from 4 public kindergartens in Nanning, and randomly divided them into experimental group(71) and control group(70). The experimental group participated in physical activities for 10 weeks (3 times a week, 1 hour each time) in the preset outdoor field (each physical activity was composed of 30 minutes of autonomous physical activity and 30 minutes of structured physical activity), while the control group had no intervention of site preset and specially designed activities. Before and after the experiment, 9 basic motor skills (including 3 fine motor skills and 6 gross motor skills) were tested.@*Results@#There were significant differences in double hand throwing, single leg balance (left), single leg balance (right), swing bridge balance and beam walking before and after intervention in experimental group(t=7.82,4.25,9.69,-10.01,-28.28,P<0.05), but there was no statistical significance in the improvement of heel toe coordinated walking(t=-0.60,P>0.05). There was no significant improvement in coin throwing (right hand), coin throwing (left hand) and square stacking(t=0.91,1.03,0.42,P>0.05), while in the control group, except for the balance of swinging bridge(t=4.27,P<0.05) and heel and toe walking(t=3.96,P<0.05), the improvement of other basic motor skills was not statistically significant(t=1.21, 0.74, 0.31, 0.97,P>0.05).@*Conclusion@#The quality of sports environment has a potential impact on the basic motor skills of preschool children. The basic motor skills of the pre-school children can be greatly improved by conducting structural and autonomous physical activities with a certain time and frequency.

15.
J Immunol ; 204(6): 1499-1507, 2020 03 15.
Article in English | MEDLINE | ID: mdl-32024699

ABSTRACT

As an important effector in response to various intracellular or extracellular stimuli, the NF-κB family extensively participates in a wide spectrum of biological events, and its dysregulation may result in many pathological conditions, such as microbial infection, tumor progression, and neurodegenerative disorders. Previous investigations showed that multiple types of ubiquitination play critical roles in the modulation of the NF-κB signaling pathway, yet the molecular mechanisms are still poorly understood. In the current study, we identified TRIM25, an E3 ubiquitin ligase, as a novel positive regulator in mediating NF-κB activation in human embryonic kidney 293T (HEK293T), HeLa cells, THP-1 cells, and PBMCs. The expression of TRIM25 promoted TNF-α-induced NF-κB signaling, whereas the knockdown had the opposite effect. Furthermore, TRIM25 interacted with TRAF2 and enhanced the K63-linked polyubiquitin chains attached to TRAF2. Moreover, TRIM25 bridged the interaction of TRAF2 and TAK1 or IKKß. To our knowledge, our study has identified a previously unrecognized role for TRIM25 in the regulation of NF-κB activation by enhancing the K63-linked ubiquitination of TRAF2.


Subject(s)
I-kappa B Kinase/metabolism , NF-kappa B/metabolism , TNF Receptor-Associated Factor 2/metabolism , Transcription Factors/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Leukocytes, Mononuclear , Lysine/metabolism , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/immunology , Primary Cell Culture , THP-1 Cells , Transcription Factors/genetics , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination/genetics , Ubiquitination/immunology
16.
Sci Adv ; 4(5): eaar2824, 2018 05.
Article in English | MEDLINE | ID: mdl-29732405

ABSTRACT

The innate immune response conferred by type I interferons is essential for host defense against viral infection but needs to be tightly controlled to avoid immunopathology. We performed a systematic functional screening by CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9) knockout and overexpression to investigate the roles of the deubiquitinating enzyme (DUB) family in regulating antiviral immunity. We demonstrated that the expression of a large fraction of DUBs underwent complex temporal alteration, suggesting a dynamic program of feedback regulation. Moreover, we identified previously unrecognized roles of a subset of DUBs, including USP5, USP14, USP22, USP48, USP52, COPS5, and BRCC3, in inhibiting antiviral immunity at various levels. We explored an unexpected mechanism where multiple DUBs, such as USP5 and USP22, form diverse signalosomes with E3 ligases or DUBs to alter the substrates' ubiquitination state instead of directly cleaving the ubiquitin chains on substrates via their protease activity. Altogether, our study has revealed a panoramic view of the broad and dynamic involvement of DUB family proteins in regulating antiviral responses.


Subject(s)
Deubiquitinating Enzymes/metabolism , Host-Pathogen Interactions , Interferon Type I/metabolism , Signal Transduction , Virus Diseases/metabolism , Deubiquitinating Enzymes/genetics , Disease Resistance/genetics , Disease Resistance/immunology , Gene Expression , Gene Expression Profiling , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Multigene Family , Proteolysis , Substrate Specificity , Ubiquitination , Virus Diseases/genetics , Virus Diseases/immunology , Virus Diseases/virology
17.
Cell Rep ; 23(3): 838-851, 2018 Apr 17.
Article in English | MEDLINE | ID: mdl-29669288

ABSTRACT

p38 signaling is broadly involved in controlling inflammation and stress-induced cell death; however, the mechanisms controlling its activity have seldom been studied. Here, we report that TRIM9 short isoform (TRIM9s) potentiates p38 signaling by stabilizing MKK6. Mechanistic studies revealed that TRIM9s promotes the K63-linked ubiquitination of MKK6 at Lys82, thus inhibiting the degradative K48-linked ubiquitination of MKK6 at the same lysine. MKK6 could also stabilize TRIM9s by promoting the phosphorylation of TRIM9s at Ser76/80 via p38, thereby blocking the ubiquitin-proteasome pathway. Further functional analyses showed that p38 signaling plays a critical role in suppressing glioblastoma progression. Co-reduction of MKK6 and TRIM9s is significantly associated with overall poor survival of glioblastoma patients. We identify a positive feedback loop in p38 signaling generated by MKK6-TRIM9s, which suppresses glioblastoma progression, and we provide insights into the mechanisms by which TRIM9s and MKK6 potentiate p38 signaling through mutual stabilization.


Subject(s)
Glioblastoma/pathology , MAP Kinase Kinase 6/metabolism , Nerve Tissue Proteins/metabolism , Signal Transduction , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Line, Tumor , Glioblastoma/drug therapy , Glioblastoma/mortality , Humans , MAP Kinase Kinase 6/deficiency , MAP Kinase Kinase 6/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Phosphorylation , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Interference , RNA, Small Interfering/metabolism , RNA, Small Interfering/therapeutic use , Survival Rate , Tripartite Motif Proteins/antagonists & inhibitors , Tripartite Motif Proteins/genetics , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics , Ubiquitination , p38 Mitogen-Activated Protein Kinases/metabolism
18.
J Mol Cell Biol ; 10(3): 205-215, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29474632

ABSTRACT

NF-κB signaling controls a large set of physiological processes ranging from inflammatory responses to cell death. Its activation is tightly regulated through controlling the activity and stability of multiple signaling components. Here, we identify that NF-κB activation is suppressed by an F-box protein, S-phase kinase associated protein 2 (SKP2). SKP2 deficiency enhanced NF-κB activation as well as the production of inflammatory cytokines. In addition, SKP2 potently blocked the NF-κB activation at the IκB kinase (IKK) level. Mechanistic study further revealed that SKP2 functions as an adaptor to promote an interaction between active IKKß and the autophagic cargo receptor p62 to mediate IKKß degradation via selective autophagy. These findings identify a previously unrecognized role of SKP2 in NF-κB activation by which SKP2 acts as a secondary receptor to assist IKKß delivery to autophagosomes for degradation in a p62-dependent manner.


Subject(s)
Autophagy , I-kappa B Kinase/immunology , NF-kappa B/immunology , S-Phase Kinase-Associated Proteins/immunology , Gene Knockout Techniques , HEK293 Cells , Humans , Proteolysis , S-Phase Kinase-Associated Proteins/genetics , Signal Transduction
19.
EMBO Rep ; 18(12): 2160-2171, 2017 12.
Article in English | MEDLINE | ID: mdl-29097393

ABSTRACT

MAVS signalosome plays an important role in RIG-I-like receptor (RLR)-induced antiviral signaling. Upon the recognition of viral RNAs, RLRs activate MAVS, which further recruits TRAF6 and other signaling proteins to initiate type I interferon (IFN) activation. MAVS signalosome also regulates virus-induced apoptosis to limit viral replication. However, the mechanisms that control the activity of MAVS signalosome are still poorly defined. Here, we report NLRP11, a Nod-like receptor, is induced by type I IFN and translocates to mitochondria to interact with MAVS upon viral infection. Using MAVS as a platform, NLRP11 degrades TRAF6 to attenuate the production of type I IFNs as well as virus-induced apoptosis. Our findings reveal the regulatory role of NLRP11 in antiviral immunity by disrupting MAVS signalosome.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis , Interferon Type I/antagonists & inhibitors , Interferon Type I/metabolism , NLR Proteins/genetics , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Humans , Immunity, Innate , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mitochondria/metabolism , NLR Proteins/metabolism , Sendai virus/physiology , Signal Transduction , THP-1 Cells , TNF Receptor-Associated Factor 6/metabolism , Virus Replication
20.
Cell Death Dis ; 8(5): e2831, 2017 05 25.
Article in English | MEDLINE | ID: mdl-28542145

ABSTRACT

Tripartite motif-containing protein 45 (TRIM45) belongs to a large family of RING-finger-containing E3 ligases, which are highly expressed in the brain. However, little is known regarding the role of TRIM45 in cancer biology, especially in human glioma. Here, we report that TRIM45 expression is significantly reduced in glioma tissue samples. Overexpression of TRIM45 suppresses proliferation and tumorigenicity in glioblastoma cells in vitro and in vivo. In addition, CRISPR/Cas9-mediated knockout of TRIM45 promotes proliferation and inhibits apoptosis in glioblastoma cells. Further mechanistic analyses show that TRIM45 interacts with and stabilizes p53. TRIM45 conjugates K63-linked polyubiquitin chain to the C-terminal six lysine residues of p53, thereby inhibiting the availability of these residues to the K48-linked polyubiquitination that targets p53 for degradation. These findings suggest that TRIM45 is a novel tumor suppressor that stabilizes and activates p53 in glioma.


Subject(s)
Brain/metabolism , Lysine/metabolism , Repressor Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Animals , Apoptosis , Biocatalysis , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Disease Progression , Down-Regulation , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/pathology , Humans , Mice, Inbred BALB C , Mice, Nude , Models, Biological , Protein Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/genetics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...