Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Cell ; 152(3): 467-78, 2013 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-23374343

RESUMO

RIG-I is a critical RNA virus sensor that serves to initiate antiviral innate immunity. However, posttranslational regulation of RIG-I signaling remains to be fully understood. We report here that RNA viruses, but not DNA viruses or bacteria, specifically upregulate lectin family member Siglecg expression in macrophages by RIG-I- or NF-κB-dependent mechanisms. Siglec-G-induced recruitment of SHP2 and the E3 ubiquitin ligase c-Cbl to RIG-I leads to RIG-I degradation via K48-linked ubiquitination at Lys813 by c-Cbl. By increasing type I interferon production, targeted inactivation of Siglecg protects mice against lethal RNA virus infection. Taken together, our data reveal a negative feedback loop of RIG-I signaling and identify a Siglec-G-mediated immune evasion pathway exploited by RNA viruses with implication in antiviral applications. These findings also provide insights into the functions and crosstalk of Siglec-G, a known adaptive response regulator, in innate immunity.


Assuntos
RNA Helicases DEAD-box/metabolismo , Infecções por Bactérias Gram-Negativas/imunologia , Imunidade Inata , Lectinas/metabolismo , Infecções por Vírus de RNA/imunologia , Receptores de Antígenos de Linfócitos B/metabolismo , Animais , Proteína DEAD-box 58 , RNA Helicases DEAD-box/química , Células Dendríticas/imunologia , Bactérias Gram-Negativas/metabolismo , Fator Regulador 3 de Interferon/metabolismo , Lectinas/genética , Lisina/metabolismo , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Proteínas Proto-Oncogênicas c-cbl/metabolismo , Vírus de RNA/metabolismo , Receptores de Antígenos de Linfócitos B/genética , Lectinas Semelhantes a Imunoglobulina de Ligação ao Ácido Siálico , Ubiquitinação
2.
Nat Immunol ; 15(7): 612-22, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24859449

RESUMO

Excessive activation of dendritic cells (DCs) leads to the development of autoimmune and inflammatory diseases, which has prompted a search for regulators of DC activation. Here we report that Rhbdd3, a member of the rhomboid family of proteases, suppressed the activation of DCs and production of interleukin 6 (IL-6) triggered by Toll-like receptors (TLRs). Rhbdd3-deficient mice spontaneously developed autoimmune diseases characterized by an increased abundance of the TH17 subset of helper T cells and decreased number of regulatory T cells due to the increase in IL-6 from DCs. Rhbdd3 directly bound to Lys27 (K27)-linked polyubiquitin chains on Lys302 of the modulator NEMO (IKKγ) via the ubiquitin-binding-association (UBA) domain in endosomes. Rhbdd3 further recruited the deubiquitinase A20 via K27-linked polyubiquitin chains on Lys268 to inhibit K63-linked polyubiquitination of NEMO and thus suppressed activation of the transcription factor NF-κB in DCs. Our data identify Rhbdd3 as a critical regulator of DC activation and indicate K27-linked polyubiquitination is a potent ubiquitin-linked pattern involved in the control of autoimmunity.


Assuntos
Proteínas Reguladoras de Apoptose/fisiologia , Autoimunidade , Células Dendríticas/imunologia , Interleucina-6/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Ubiquitinação , Animais , Interleucina-6/antagonistas & inibidores , Lisina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/fisiologia , Estrutura Terciária de Proteína , Linfócitos T/imunologia , Receptores Toll-Like/fisiologia
3.
Brain ; 147(7): 2552-2565, 2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38366606

RESUMO

Chronic varicella zoster virus (VZV) infection induced neuroinflammatory condition is the critical pathology of post-herpetic neuralgia (PHN). The immune escape mechanism of VZV remains elusive. As to mice have no VZV infection receptor, herpes simplex virus type 1 (HSV-1) infection is a well established PHN mice model. Transcriptional expression analysis identified that the protein arginine methyltransferases 6 (Prmt6) was upregulated upon HSV-1 infection, which was further confirmed by immunofluorescence staining in spinal dorsal horn. Prmt6 deficiency decreased HSV-1-induced neuroinflammation and PHN by enhancing antiviral innate immunity and decreasing HSV-1 load in vivo and in vitro. Overexpression of Prmt6 in microglia dampened antiviral innate immunity and increased HSV-1 load. Mechanistically, Prmt6 methylated and inactivated STING, resulting in reduced phosphorylation of TANK binding kinase-1 (TBK1) and interferon regulatory factor 3 (IRF3), diminished production of type I interferon (IFN-I) and antiviral innate immunity. Furthermore, intrathecal or intraperitoneal administration of the Prmt6 inhibitor EPZ020411 decreased HSV-1-induced neuroinflammation and PHN by enhancing antiviral innate immunity and decreasing HSV-1 load. Our findings revealed that HSV-1 escapes antiviral innate immunity and results in PHN by upregulating Prmt6 expression and inhibiting the cGAS-STING pathway, providing novel insights and a potential therapeutic target for PHN.


Assuntos
Herpesvirus Humano 1 , Proteínas de Membrana , Neuralgia Pós-Herpética , Nucleotidiltransferases , Proteína-Arginina N-Metiltransferases , Regulação para Cima , Animais , Proteína-Arginina N-Metiltransferases/metabolismo , Proteína-Arginina N-Metiltransferases/genética , Proteína-Arginina N-Metiltransferases/antagonistas & inibidores , Camundongos , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Nucleotidiltransferases/metabolismo , Nucleotidiltransferases/genética , Neuralgia Pós-Herpética/metabolismo , Neuralgia Pós-Herpética/imunologia , Camundongos Endogâmicos C57BL , Imunidade Inata , Humanos , Camundongos Knockout , Masculino , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 3 de Interferon/genética , Herpes Simples/imunologia , Microglia/metabolismo , Microglia/imunologia , Proteínas Serina-Treonina Quinases
4.
Nat Immunol ; 13(6): 551-9, 2012 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-22522491

RESUMO

The molecular mechanisms that fine-tune Toll-like receptor (TLR)-triggered innate inflammatory responses remain to be fully elucidated. Major histocompatibility complex (MHC) molecules can mediate reverse signaling and have nonclassical functions. Here we found that constitutively expressed membrane MHC class I molecules attenuated TLR-triggered innate inflammatory responses via reverse signaling, which protected mice from sepsis. The intracellular domain of MHC class I molecules was phosphorylated by the kinase Src after TLR activation, then the tyrosine kinase Fps was recruited via its Src homology 2 domain to phosphorylated MHC class I molecules. This led to enhanced Fps activity and recruitment of the phosphatase SHP-2, which interfered with TLR signaling mediated by the signaling molecule TRAF6. Thus, constitutive MHC class I molecules engage in crosstalk with TLR signaling via the Fps-SHP-2 pathway and control TLR-triggered innate inflammatory responses.


Assuntos
Antígenos de Histocompatibilidade Classe I/imunologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/imunologia , Proteínas Proto-Oncogênicas c-fes/imunologia , Receptores Toll-Like/imunologia , Animais , Escherichia coli/imunologia , Imunidade Inata/imunologia , Immunoblotting , Interferon beta/imunologia , Interleucina-6/imunologia , Listeria monocytogenes/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Transdução de Sinais/imunologia , Fator de Necrose Tumoral alfa/imunologia
5.
Brain Behav Immun ; 118: 101-114, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38402915

RESUMO

Microglia induced chronic inflammation is the critical pathology of Neuropathic pain (NP). Metabolic reprogramming of macrophage has been intensively reported in various chronic inflammation diseases. However, the metabolic reprogramming of microglia in chronic pain remains to be elusive. Here, we reported that immuno-metabolic markers (HIF-1α, PKM2, GLUT1 and lactate) were related with increased expression of PRMT6 in the ipsilateral spinal cord dorsal horn of the chronic construction injury (CCI) mice. PRMT6 deficiency or prophylactic and therapeutic intrathecal administration of PRMT6 inhibitor (EPZ020411) ameliorated CCI-induced NP, inflammation and glycolysis in the ipsilateral spinal cord dorsal horn. PRMT6 knockout or knockdown inhibited LPS-induced inflammation, proliferation and glycolysis in microglia cells. While PRMT6 overexpression exacerbated LPS-induced inflammation, proliferation and glycolysis in BV2 cells. Recent research revealed that PRMT6 could interact with and methylate HIF-1α, which increased HIF-1α protein stability. In sum, increased expression of PRMT6 exacerbates NP progress by increasing glycolysis and neuroinflammation through interacting with and stabilizing HIF-1α in a methyltransferase manner, which outlines novel pathological mechanism and drug target for NP.


Assuntos
Microglia , Neuralgia , Camundongos , Animais , Microglia/metabolismo , Lipopolissacarídeos/farmacologia , Lipopolissacarídeos/metabolismo , Inflamação/metabolismo , Neuralgia/metabolismo , Glicólise
6.
Pharmacol Res ; 201: 107096, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38320736

RESUMO

The uncontrolled bacterial infection-induced cytokine storm and sequential immunosuppression are commonly observed in septic patients, which indicates that the activation of phagocytic cells and the efficient and timely elimination of bacteria are crucial for combating bacterial infections. However, the role of dysregulated immune cells and their disrupted function in sepsis remains unclear. Here, we found that macrophages exhibited the impaired endocytosis capabilities in sepsis by Single-cell RNA sequencing and bulk RNA sequencing. Caveolae protein Caveolin-1 (Cav-1) of macrophages was inactivated by SHP2 rapidly during Escherichia coli (E.coli) infection. Allosteric inhibitor of SHP2 effectively maintains Cav-1 phosphorylation to enhance macrophage to endocytose and eliminate bacteria. Additionally, TLR4 endocytosis of macrophage was also enhanced upon E.coli infection by SHP099, inducing an increased and rapidly resolved inflammatory response. In vivo, pretreatment or posttreatment with inhibitor of SHP2 significantly reduced the bacterial burden in organs and mortality of mice subjected E.coli infection or CLP-induced sepsis. The cotreatment of inhibitor of SHP2 with an antibiotic conferred complete protection against mortality in mice. Our findings suggest that Cav-1-mediated endocytosis and bacterial elimination may play a critical role in the pathogenesis of sepsis, highlighting inhibitor of SHP2 as a potential therapeutic agent for sepsis.


Assuntos
Cavéolas , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Sepse , Animais , Humanos , Camundongos , Bactérias , Cavéolas/metabolismo , Endocitose , Infecções por Escherichia coli/tratamento farmacológico , Infecções por Escherichia coli/metabolismo , Macrófagos , Proteína Tirosina Fosfatase não Receptora Tipo 11/antagonistas & inibidores , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Sepse/tratamento farmacológico , Sepse/metabolismo
7.
J Integr Neurosci ; 23(4): 82, 2024 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-38682225

RESUMO

BACKGROUND: Comorbid chronic neuropathic pain (NPP) and anxio-depressive disorders (ADD) have become a serious global public-health problem. The SLIT and NTRK-like 1 (SLITRK1) protein is important for synaptic remodeling and is highly expressed in the amygdala, an important brain region involved in various emotional behaviors. We examined whether SLITRK1 protein in the amygdala participates in NPP and comorbid ADD. METHODS: A chronic NPP mouse model was constructed by L5 spinal nerve ligation; changes in chronic pain and ADD-like behaviors were measured in behavioral tests. Changes in SLITRK1 protein and excitatory synaptic functional proteins in the amygdala were measured by immunofluorescence and Western blot. Adeno-associated virus was transfected into excitatory synaptic neurons in the amygdala to up-regulate the expression of SLITRK1. RESULTS: Chronic NPP-related ADD-like behavior was successfully produced in mice by L5 ligation. We found that chronic NPP and related ADD decreased amygdalar expression of SLITRK1 and proteins important for excitatory synaptic function, including Homer1, postsynaptic density protein 95 (PSD95), and synaptophysin. Virally-mediated SLITRK1 overexpression in the amygdala produced a significant easing of chronic NPP and ADD, and restored the expression levels of Homer1, PSD95, and synaptophysin. CONCLUSION: Our findings indicated that SLITRK1 in the amygdala plays an important role in chronic pain and related ADD, and may prove to be a potential therapeutic target for chronic NPP-ADD comorbidity.


Assuntos
Tonsila do Cerebelo , Comportamento Animal , Dor Crônica , Proteína 4 Homóloga a Disks-Large , Proteínas do Tecido Nervoso , Neuralgia , Animais , Masculino , Camundongos , Tonsila do Cerebelo/metabolismo , Ansiedade/metabolismo , Ansiedade/fisiopatologia , Transtornos de Ansiedade/metabolismo , Transtornos de Ansiedade/fisiopatologia , Comportamento Animal/fisiologia , Dor Crônica/metabolismo , Dor Crônica/fisiopatologia , Depressão/metabolismo , Depressão/etiologia , Depressão/fisiopatologia , Transtorno Depressivo/metabolismo , Transtorno Depressivo/fisiopatologia , Modelos Animais de Doenças , Proteína 4 Homóloga a Disks-Large/metabolismo , Proteínas de Arcabouço Homer/metabolismo , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/metabolismo , Neuralgia/metabolismo , Sinaptofisina/metabolismo
8.
Mol Cancer ; 21(1): 164, 2022 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-35974394

RESUMO

Developing a strategy to specifically kill cancer cells without inducing obvious damage to normal cells may be of great clinical significance for cancer treatment. In the present study, we developed a new precise personalized strategy named "i-CRISPR" for cancer treatment through adding DNA damage repair inhibitors(i) and inducing cancer cell-specific DNA double strand breaks by CRISPR. Through in vitro and in vivo experiments, we confirmed the efficacy of this strategy in multiple cancer models and revealed the mechanism of cell death. Our strategy might provide a novel concept for precise cancer therapy.


Assuntos
Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Neoplasias , Sistemas CRISPR-Cas , Quebras de DNA de Cadeia Dupla , Edição de Genes , Humanos , Mutação , Neoplasias/genética , Neoplasias/terapia
9.
Nat Immunol ; 11(8): 734-42, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20639876

RESUMO

Integrins are critical for the migration and function of leukocytes in inflammation. However, the interaction between integrin alpha(M) (CD11b), which has high expression in monocytes and macrophages, and Toll-like receptor (TLR)-triggered innate immunity remains unclear. Here we report that CD11b deficiency enhanced TLR-mediated responses in macrophages, rendering mice more susceptible to endotoxin shock and Escherichia coli-caused sepsis. CD11b was activated by TLR-triggered phosphatidylinositol 3-OH kinase (PI(3)K) and the effector RapL and fed back to inhibit TLR signaling by activating the tyrosine kinases Src and Syk. Syk interacted with and induced tyrosine phosphorylation of MyD88 and TRIF, which led to degradation of these adaptor molecules by the E3 ubiquitin ligase Cbl-b. Thus, TLR-triggered, active CD11b integrin engages in crosstalk with the MyD88 and TRIF pathways and subsequently inhibits TLR signaling in innate immune responses.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/imunologia , Antígeno CD11b/imunologia , Inflamação/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Proteínas Tirosina Quinases/imunologia , Proteínas Proto-Oncogênicas c-cbl/imunologia , Receptores Toll-Like/imunologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/imunologia , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Ativação Enzimática , Escherichia coli/imunologia , Infecções por Escherichia coli/imunologia , Regulação Enzimológica da Expressão Gênica , Imunidade Inata/imunologia , Inflamação/enzimologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Macrófagos/imunologia , Camundongos , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/imunologia , Fator 88 de Diferenciação Mieloide/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-cbl/metabolismo , Transdução de Sinais , Quinase Syk , Quinases da Família src/imunologia
10.
Immunity ; 39(3): 470-81, 2013 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-24012418

RESUMO

Histone modifications play important roles in multiple physiological processes by regulating gene expression. However, the roles of histone modifications in immunity remain poorly understood. Here we report that Ash1l, a H3K4 methyltransferase, suppressed interleukin-6 (IL-6), and tumor necrosis factor (TNF) production in Toll-like receptor (TLR)-triggered macrophages, protecting mice from sepsis. Ash1l-silenced mice were more susceptible to autoimmune disease as a result of enhanced IL-6 production. Ash1l enhanced A20 expression through induction of H3K4 modification at the Tnfaip3 promoter via H3K4 methyltransferase activity of Ash1l SET (Su[var]3-9, E[z] and trithorax) domain. Ash1l suppressed NF-κB, mitogen-activated protein kinase (MAPK) pathways, and subsequent IL-6 production via facilitating A20-mediated NF-κB signal modulator NEMO and transducer TRAF6 deubiquitination. Therefore, Ash1l-mediated H3K4 methylation at the Tnfaip3 promoter is required for controlling innate IL-6 production and suppressing inflammatory autoimmune diseases, providing mechanistic insight into epigenetic modulation of immune responses and inflammation.


Assuntos
Doenças Autoimunes/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas de Ligação a DNA/metabolismo , Interleucina-6/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Células Cultivadas , Cisteína Endopeptidases , Proteínas de Ligação a DNA/biossíntese , Histonas/metabolismo , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Inflamação , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Macrófagos , Camundongos , Camundongos Knockout , NF-kappa B/metabolismo , Regiões Promotoras Genéticas , Interferência de RNA , RNA Interferente Pequeno , Sepse/imunologia , Sepse/prevenção & controle , Transdução de Sinais , Fator 6 Associado a Receptor de TNF/metabolismo , Receptores Toll-Like/metabolismo , Proteína 3 Induzida por Fator de Necrose Tumoral alfa , Fatores de Necrose Tumoral/biossíntese , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/biossíntese
11.
Proc Natl Acad Sci U S A ; 116(26): 12958-12963, 2019 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-31189603

RESUMO

Macrophages can internalize the invading pathogens by raft/caveolae and/or clathrin-dependent endocytosis and elicit an immune response against infection. However, the molecular mechanism for macrophage endocytosis remains elusive. Here we report that LAPF (lysosome-associated and apoptosis-inducing protein containing PH and FYVE domains) is required for caveolae-mediated endocytosis. Lapf-deficient macrophages have impaired capacity to endocytose and eliminate bacteria. Macrophage-specific Lapf-deficient mice are more susceptible to Escherichia coli (E. coli) infection with higher bacterial loads. Moreover, Lapf deficiency impairs TLR4 endocytosis, resulting in attenuated production of TLR-triggered proinflammatory cytokines. LAPF is localized to early endosomes and interacts with caveolin-1. Phosphorylation of LAPF by the tyrosine kinase Src is required for LAPF-Src-Caveolin complex formation and endocytosis and elimination of bacteria. Collectively, our work demonstrates that LAPF is critical for endocytosis of bacteria and induction of inflammatory responses, suggesting that LAPF and Src could be potential targets for the control of infectious diseases.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Caveolina 1/metabolismo , Endocitose/imunologia , Infecções por Escherichia coli/imunologia , Macrófagos/imunologia , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/imunologia , Caveolina 1/imunologia , Linhagem Celular , Citocinas/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Endossomos/imunologia , Endossomos/metabolismo , Endossomos/microbiologia , Escherichia coli/imunologia , Infecções por Escherichia coli/microbiologia , Imunidade Inata , Lipopolissacarídeos/administração & dosagem , Lipopolissacarídeos/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , Camundongos Knockout , Cultura Primária de Células , Quinases da Família src/imunologia , Quinases da Família src/metabolismo
12.
Cytokine ; 111: 209-215, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30176559

RESUMO

The balance between pro-inflammatory and anti-inflammatory macrophage generation, a process known as polarization, is critical for immune homoeostasis. Identifying the molecular mechanisms underlying polarization and the generation of anti-inflammatory macrophages is an area of high current interest. Our previous work has demonstrated that integrin CD11b promotes IL-10 production in macrophages by activating the Sarcoma gene (Src), yet whether and how Src modulates anti-inflammatory macrophages is not known. Here we show that Src inhibitor (Dasatinib)-treated mice were highly susceptible to dextran sulfate sodium (DSS)-induced colitis, with a much more sever inflammation response, accompanying by high TNF-α, and low IL-10 expression. Inhibition of Src enhanced the expression of pro-inflammatory macrophage marker inducible nitric oxide synthase (iNOS), but reduced the expression of anti-inflammatory macrophage marker arginase1 (Arg1) in both intestinal macrophages and bone marrow-derived macrophages (BMDMs). Overexpression of constitutively activated Src promoted IL-4-induced expression of Arg1 through STAT6 phosphorylation, and Jak1 was also involved in this process. Our results reveal that the IL-4-Src-STAT6 pathway plays a major role in polarizing anti-inflammatory macrophage generation and suggest a potential anti-inflammatory role for Src in inflammatory diseases.


Assuntos
Anti-Inflamatórios/metabolismo , Interleucina-4/metabolismo , Macrófagos/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Fator de Transcrição STAT6/metabolismo , Animais , Linhagem Celular , Colite/metabolismo , Células HEK293 , Humanos , Interleucina-10/metabolismo , Janus Quinase 1/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo II/metabolismo , Células RAW 264.7 , Transdução de Sinais/fisiologia , Fator de Necrose Tumoral alfa/metabolismo
14.
J Autoimmun ; 81: 120-129, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28461090

RESUMO

The induction and persistence of a hypo-inflammatory and immunosuppressive state in severe sepsis is commonly associated with increased risks of secondary infections and mortality. Toll-like receptor (TLR)-triggered inflammatory response of macrophages/monocytes plays an important role in determining the outcome of hyper-inflammation during the acute phase and the hypo-inflammation during immunosuppressive phase of sepsis. However, the mechanisms for controlling hypo-inflammatory response in endotoxin tolerant macrophages remain to be fully understood. Considering that metabolic control of inflammation is an emerging field and the balance between AMP/ATP and oxidized NAD+/reduced NADH is associated with inflammation and metabolism, we analyzed the level of NAD+ in TLR-triggered innate inflammatory response, and found that the decreased level of NAD+ was significantly related to the increased inflammatory cytokine production both in vivo and in vitro. By screening the expression and function of NAD+ dependent type III deacetylase Sirtuin family members, we found that SIRT5 and SIRT1/2 had opposite expression patterns and functions in macrophages. SIRT5 deficiency decreased TLR-triggered inflammation in both acute and immunosuppressive phases of sepsis. Interestingly, cytoplasmic SIRT5 counteracted the inhibitory effects of SIRT2 and enhanced the innate inflammatory responses in macrophages and even in endotoxin-tolerant macrophages by promoting acetylation of p65 and activation of NF-κB pathway. Mechanistically, SIRT5 competed with SIRT2 to interact with NF-κB p65, in a deacetylase activity-independent way, to block the deacetylation of p65 by SIRT2, which consequently led to increased acetylation of p65 and the activation of NF-κB pathway and its downstream cytokines. Our study discovered the new functions of different Sirtuin members in sepsis, indicating that targeting of Sirtuin family members at different sepsis phases can be helpful to precisely control the progression of sepsis.


Assuntos
Endotoxinas/imunologia , Tolerância Imunológica , Inflamação/imunologia , Inflamação/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , NAD/metabolismo , Sirtuínas/metabolismo , Fator de Transcrição RelA/metabolismo , Acetilação , Animais , Linhagem Celular , Citocinas/metabolismo , Expressão Gênica , Genótipo , Humanos , Imunidade Inata , Imunomodulação , Mediadores da Inflamação/metabolismo , Camundongos , Camundongos Knockout
16.
J Autoimmun ; 69: 74-85, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26994903

RESUMO

The migration of Th17 cells into central nervous system (CNS) tissue is the key pathogenic step in experimental autoimmune encephalomyelitis (EAE) model. However, the mechanism underlying the pathogenic Th17 cell migration remains elusive. Here we report that blockade of CD47 with CD47-Fc fusion protein is effective in preventing and curing EAE by impairing infiltration of Th17 cells into CNS. However, CD47 deficiency does not directly impair the migration of Th17 cells. Mechanistic studies showed that CD47 deficiency inhibited degradation of inducible nitric oxide synthase (iNOS) in proteasome of macrophages by Src activation and led to the increased nitric oxide (NO) production. Then NO suppressed inflammasome activation-induced IL-1ß production. This lower IL-1ß reduces the expression of IL-1R1 and migration-related chemokine receptors on CD47(-/-) Th17 cells, inhibiting the ability of Th17 cells to infiltrate into the CNS of CD47(-/-) mice and therefore suppressing EAE development. In vivo administration of exogenous IL-1ß indeed promoted the infiltration CD47(-/-) Th17 cells into CNS and antagonized the protective role of CD47 deficiency in EAE pathogenesis. Our results demonstrate a potential preventive and therapeutic application of CD47 blockade in controlling EAE development.


Assuntos
Autoimunidade , Antígeno CD47/metabolismo , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Interleucina-1/metabolismo , Células Th17/imunologia , Células Th17/metabolismo , Animais , Anticorpos Bloqueadores/farmacologia , Anticorpos Monoclonais/farmacologia , Células da Medula Óssea , Antígeno CD47/genética , Movimento Celular/genética , Movimento Celular/imunologia , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/patologia , Fragmentos Fc das Imunoglobulinas/genética , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Esclerose Múltipla/genética , Esclerose Múltipla/imunologia , Esclerose Múltipla/metabolismo , Esclerose Múltipla/patologia , Óxido Nítrico Sintase Tipo II/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Recombinantes de Fusão , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Quinases da Família src/metabolismo
17.
J Biol Chem ; 288(23): 16225-16234, 2013 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-23609450

RESUMO

Sepsis-associated immunosuppression (SAIS) is regarded as one of main causes for the death of septic patients at the late stage because of the decreased innate immunity with a more opportunistic infection. LPS-tolerized macrophages, which are re-challenged by LPS after prior exposure to LPS, are regarded as the common model of hypo-responsiveness for SAIS. However, the molecular mechanisms of endotoxin tolerance and SAIS remain to be fully elucidated. In addition, negative regulation of the Toll-like receptor (TLR)-triggered innate inflammatory response needs further investigation. Here we show that expression of immune responsive gene 1 (IRG1) was highly up-regulated in the peripheral blood mononuclear cells of septic patients and in LPS-tolerized mouse macrophages. IRG1 significantly suppressed TLR-triggered production of proinflammatory cytokines TNF-α, IL-6, and IFN-ß in LPS-tolerized macrophages, with the elevated expression of reactive oxygen species (ROS) and A20. Moreover, ROS enhanced A20 expression by increasing the H3K4me3 modification of histone on the A20 promoter domain, and supplement of the ROS abrogated the IRG1 knockdown function in breaking endotoxin tolerance by increasing A20 expression. Our results demonstrate that inducible IRG1 promotes endotoxin tolerance by increasing A20 expression through ROS, indicating a new molecular mechanism regulating hypoinflammation of sepsis and endotoxin tolerance.


Assuntos
Proteínas de Ligação a DNA/imunologia , Regulação da Expressão Gênica/efeitos dos fármacos , Hidroliases/imunologia , Tolerância Imunológica/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Lipopolissacarídeos/toxicidade , Macrófagos/imunologia , Proteínas Nucleares/imunologia , Proteínas/imunologia , Espécies Reativas de Oxigênio/imunologia , Sepse/imunologia , Ubiquitina-Proteína Ligases/imunologia , Animais , Carboxiliases , Cisteína Endopeptidases , Proteínas de Ligação a DNA/genética , Feminino , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Técnicas de Silenciamento de Genes , Histonas/genética , Histonas/imunologia , Humanos , Hidroliases/genética , Interferon beta/genética , Interferon beta/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Lipopolissacarídeos/imunologia , Macrófagos/patologia , Masculino , Camundongos , Monócitos/imunologia , Monócitos/patologia , Proteínas Nucleares/genética , Proteínas/genética , Sepse/genética , Sepse/patologia , Proteína 3 Induzida por Fator de Necrose Tumoral alfa , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia , Ubiquitina-Proteína Ligases/genética
18.
Cell Rep ; 43(5): 114135, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38652662

RESUMO

Optimal activation of stimulator of interferon genes (STING) protein is crucial for host defenses against pathogens and avoiding detrimental effects. Various post-translational modifications control STING activity. However, the function of interferon (IFN)-stimulated gene (ISG) 15 modification (ISGylation) in controlling STING stability and activation is unclear. Here, we show that the E3 ISGylation ligases HECT domain- and RCC1-like domain-containing proteins (HERCs; HERC5 in humans and HERC6 in mice) facilitate STING activation by mediating ISGylation of STING at K150, preventing its K48-linked ubiquitination and degradation. Concordantly, Herc6 deficiency suppresses herpes simplex virus 1 infection-induced type I IFN responses and facilitates viral replication both in vitro and in vivo. Notably, severe acute respiratory syndrome coronavirus 2 protein papain-like protease cleaves HERC5-mediated ISGylation of STING, suppressing host antiviral responses. These data identify a mechanism by which HERCs-mediated ISGylation controls STING stability and activation and uncover the correlations and interactions of ISGylation and ubiquitination during STING activation.


Assuntos
Proteínas de Membrana , Ubiquitina-Proteína Ligases , Ubiquitinação , Ubiquitinas , Animais , Humanos , Camundongos , Citocinas/metabolismo , Células HEK293 , Herpes Simples/virologia , Herpes Simples/metabolismo , Herpes Simples/imunologia , Herpesvirus Humano 1/fisiologia , Interferon Tipo I/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , SARS-CoV-2/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinas/metabolismo , Replicação Viral , Masculino , Feminino
19.
CNS Neurosci Ther ; 30(8): e14913, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39123294

RESUMO

BACKGROUND: Hyperglycemia-induced neuroinflammation significantly contributes to diabetic neuropathic pain (DNP), but the underlying mechanisms remain unclear. OBJECTIVE: To investigate the role of Sirt3, a mitochondrial deacetylase, in hyperglycemia-induced neuroinflammation and DNP and to explore potential therapeutic interventions. METHOD AND RESULTS: Here, we found that Sirt3 was downregulated in spinal dorsal horn (SDH) of diabetic mice by RNA-sequencing, which was further confirmed at the mRNA and protein level. Sirt3 deficiency exacerbated hyperglycemia-induced neuroinflammation and DNP by enhancing microglial aerobic glycolysis in vivo and in vitro. Overexpression of Sirt3 in microglia alleviated inflammation by reducing aerobic glycolysis. Mechanistically, high-glucose stimulation activated Akt, which phosphorylates and inactivates FoxO1. The inactivation of FoxO1 diminished the transcription of Sirt3. Besides that, we also found that hyperglycemia induced Sirt3 degradation via the mitophagy-lysosomal pathway. Blocking Akt activation by GSK69093 or metformin rescued the degradation of Sirt3 protein and transcription inhibition of Sirt3 mRNA, which substantially diminished hyperglycemia-induced inflammation. Metformin in vivo treatment alleviated neuroinflammation and diabetic neuropathic pain by rescuing hyperglycemia-induced Sirt3 downregulation. CONCLUSION: Hyperglycemia induces metabolic reprogramming and inflammatory activation in microglia through the regulation of Sirt3 transcription and degradation. This novel mechanism identifies Sirt3 as a potential drug target for treating DNP.


Assuntos
Diabetes Mellitus Experimental , Neuropatias Diabéticas , Regulação para Baixo , Glicólise , Hiperglicemia , Camundongos Endogâmicos C57BL , Microglia , Sirtuína 3 , Animais , Sirtuína 3/metabolismo , Sirtuína 3/genética , Camundongos , Glicólise/efeitos dos fármacos , Glicólise/fisiologia , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Hiperglicemia/metabolismo , Microglia/metabolismo , Microglia/efeitos dos fármacos , Masculino , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/complicações , Neuropatias Diabéticas/metabolismo , Inflamação/metabolismo , Doenças Neuroinflamatórias/metabolismo , Doenças Neuroinflamatórias/etiologia , Metformina/farmacologia
20.
Adv Sci (Weinh) ; : e2403177, 2024 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-39120025

RESUMO

Epigenetic regulation of metabolism profoundly influences cell fate commitment. During osteoclast differentiation, the activation of RANK signaling is accompanied by metabolic reprogramming, but the epigenetic mechanisms by which RANK signaling induces this reprogramming remain elusive. By transcriptional sequence and ATAC analysis, this study identifies that activation of RANK signaling upregulates PRMT6 by epigenetic modification, triggering a metabolic switching from fatty acids oxidation toward glycolysis. Conversely, Prmt6 deficiency reverses this shift, markedly reducing HIF-1α-mediated glycolysis and enhancing fatty acid oxidation. Consequently, PRMT6 deficiency or inhibitor impedes osteoclast differentiation and alleviates bone loss in ovariectomized (OVX) mice. At the molecular level, Prmt6 deficiency reduces asymmetric dimethylation of H3R2 at the promoters of genes including Ppard, Acox3, and Cpt1a, enhancing genomic accessibility for fatty acid oxidation. PRMT6 thus emerges as a metabolic checkpoint, mediating metabolic switch from fatty acid oxidation to glycolysis, thereby supporting osteoclastogenesis. Unveiling PRMT6's critical role in epigenetically orchestrating metabolic shifts in osteoclastogenesis offers a promising target for anti-resorptive therapy.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA