Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 150
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(1)2022 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-34930823

RESUMO

Coxiella burnetii is a bacterial pathogen that replicates within host cells by establishing a membrane-bound niche called the Coxiella-containing vacuole. Biogenesis of this compartment requires effectors of its Dot/Icm type IV secretion system. A large cohort of such effectors has been identified, but the function of most of them remain elusive. Here, by a cell-based functional screening, we identified the effector Cbu0513 (designated as CinF) as an inhibitor of NF-κB signaling. CinF is highly similar to a fructose-1,6-bisphosphate (FBP) aldolase/phosphatase present in diverse bacteria. Further study reveals that unlike its ortholog from Sulfolobus tokodaii, CinF does not exhibit FBP phosphatase activity. Instead, it functions as a protein phosphatase that specifically dephosphorylates and stabilizes IκBα. The IκBα phosphatase activity is essential for the role of CinF in C. burnetii virulence. Our results establish that C. burnetii utilizes a protein adapted from sugar metabolism to subvert host immunity.


Assuntos
Proteínas de Bactérias , Coxiella burnetii , Fosfoproteínas Fosfatases , Febre Q , Transdução de Sinais , Fatores de Virulência , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Chlorocebus aethiops , Coxiella burnetii/genética , Coxiella burnetii/imunologia , Coxiella burnetii/patogenicidade , Células HEK293 , Células HeLa , Humanos , NF-kappa B/genética , NF-kappa B/imunologia , Fosfoproteínas Fosfatases/genética , Fosfoproteínas Fosfatases/imunologia , Febre Q/genética , Febre Q/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Células Vero , Fatores de Virulência/genética , Fatores de Virulência/imunologia
2.
J Neuroinflammation ; 18(1): 145, 2021 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-34183017

RESUMO

BACKGROUND: Experimental autoimmune encephalomyelitis (EAE) is an animal disease model of multiple sclerosis (MS) that involves the immune system and central nervous system (CNS). However, it is unclear how genetic predispositions promote neuroinflammation in MS and EAE. Here, we investigated how partial loss-of-function of suppressor of MEK1 (SMEK1), a regulatory subunit of protein phosphatase 4, facilitates the onset of MS and EAE. METHODS: C57BL/6 mice were immunized with myelin oligodendrocyte glycoprotein 35-55 (MOG35-55) to establish the EAE model. Clinical signs were recorded and pathogenesis was investigated after immunization. CNS tissues were analyzed by immunostaining, quantitative polymerase chain reaction (qPCR), western blot analysis, and enzyme-linked immunosorbent assay (ELISA). Single-cell analysis was carried out in the cortices and hippocampus. Splenic and lymph node cells were evaluated with flow cytometry, qPCR, and western blot analysis. RESULTS: Here, we showed that partial Smek1 deficiency caused more severe symptoms in the EAE model than in controls by activating myeloid cells and that Smek1 was required for maintaining immunosuppressive function by modulating the indoleamine 2,3-dioxygenase (IDO1)-aryl hydrocarbon receptor (AhR) pathway. Single-cell sequencing and an in vitro study showed that Smek1-deficient microglia and macrophages were preactivated at steady state. After MOG35-55 immunization, microglia and macrophages underwent hyperactivation and produced increased IL-1ß in Smek1-/+ mice at the peak stage. Moreover, dysfunction of the IDO1-AhR pathway resulted from the reduction of interferon γ (IFN-γ), enhanced antigen presentation ability, and inhibition of anti-inflammatory processes in Smek1-/+ EAE mice. CONCLUSIONS: The present study suggests a protective role of Smek1 in autoimmune demyelination pathogenesis via immune suppression and inflammation regulation in both the immune system and the central nervous system. Our findings provide an instructive basis for the roles of Smek1 in EAE and broaden the understanding of the genetic factors involved in the pathogenesis of autoimmune demyelination.


Assuntos
Sistema Nervoso Central/patologia , Encefalomielite Autoimune Experimental , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Interferon gama/metabolismo , Microglia/imunologia , Fosfoproteínas Fosfatases/imunologia , Receptores de Hidrocarboneto Arílico/metabolismo , Animais , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/fisiopatologia , Citocinas , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Técnicas de Inativação de Genes , Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Esclerose Múltipla/imunologia , Glicoproteína Mielina-Oligodendrócito/imunologia , Células Mieloides/imunologia , Células Mieloides/metabolismo , Fragmentos de Peptídeos/imunologia , Fosfoproteínas Fosfatases/metabolismo , Transdução de Sinais , Baço/patologia
3.
Clin Exp Immunol ; 203(1): 41-46, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32979862

RESUMO

During a 15-year period, the incidence of type 1 diabetes has doubled in Lithuania, while increasing by a third in England; however, England still has a higher incidence. Analysis of sera collected from non-diabetic schoolchildren from Lithuania and England more than 20 years ago showed a similar number of multiple autoantibody-positive schoolchildren between the populations, but a higher prevalence of islet antigen-2 autoantibodies (IA-2A) in English schoolchildren. We aimed to use recently developed, more specific islet autoantibody tests to characterize differences in humoral autoimmunity between these two general population cohorts in greater detail. Samples from 88 Lithuanian and 133 English schoolchildren previously found islet autoantibody-positive were selected for measurement of additional islet autoantibodies by radioimmunoassay. Samples were tested for autoantibodies to zinc transporter 8 (ZnT8A), GAD (96-585), the protein tyrosine phosphatase region of islet antigen-2 (PTPA) and the related IA-2ßA, while autoantibodies to IA-2A were reassayed using the current harmonized method. IA-2-related autoantibodies PTPA (0·13 versus 0·45%, P = 0·027) and IA-2ßA (0 versus 0·35%, P < 0·001), but not IA-2A measured using the harmonized method, were less common in Lithuanian compared to English schoolchildren. Lithuanian schoolchildren who were islet autoantibody-positive were positive for fewer biochemical autoantibodies compared with English schoolchildren (P = 0·043). Background rates of islet autoimmunity in childhood differ subtly between countries, which have different incidences of type 1 diabetes. The optimal screening strategy (age and combination of markers) for detection of islet autoimmunity may vary between countries, dependent upon the pattern of autoantibodies found in the general population.


Assuntos
Autoanticorpos/sangue , Diabetes Mellitus Tipo 1/sangue , Ilhotas Pancreáticas/metabolismo , Adolescente , Autoanticorpos/imunologia , Criança , Pré-Escolar , Diabetes Mellitus Tipo 1/imunologia , Inglaterra , Feminino , Glutamato Descarboxilase/imunologia , Glutamato Descarboxilase/metabolismo , Humanos , Ilhotas Pancreáticas/imunologia , Lituânia , Masculino , Fosfoproteínas Fosfatases/imunologia , Fosfoproteínas Fosfatases/metabolismo , Transportador 8 de Zinco/imunologia , Transportador 8 de Zinco/metabolismo
4.
mBio ; 11(4)2020 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-32753499

RESUMO

Stimulator of interferon genes (STING) is an essential adaptor protein of the innate DNA-sensing signaling pathway, which recognizes genomic DNA from invading pathogens to establish antiviral responses in host cells. STING activity is tightly regulated by several posttranslational modifications, including phosphorylation. However, specifically how the phosphorylation status of STING is modulated by kinases and phosphatases remains to be fully elucidated. In this study, we identified protein phosphatase 6 catalytic subunit (PPP6C) as a binding partner of Kaposi's sarcoma-associated herpesvirus (KSHV) open reading frame 48 (ORF48), which is a negative regulator of the cyclic GMP-AMP synthase (cGAS)-STING pathway. PPP6C depletion enhances double-stranded DNA (dsDNA)-induced and 5'ppp double-stranded RNA (dsRNA)-induced but not poly(I:C)-induced innate immune responses. PPP6C negatively regulates dsDNA-induced IRF3 activation but not NF-κB activation. Deficiency of PPP6C greatly inhibits the replication of herpes simplex virus 1 (HSV-1) and vesicular stomatitis virus (VSV) as well as the reactivation of KSHV, due to increased type I interferon production. We further demonstrated that PPP6C interacts with STING and that loss of PPP6C enhances STING phosphorylation. These data demonstrate the important role of PPP6C in regulating STING phosphorylation and activation, which provides an additional mechanism by which the host responds to viral infection.IMPORTANCE Cytosolic DNA, which usually comes from invading microbes, is a dangerous signal to the host. The cGAS-STING pathway is the major player that detects cytosolic DNA and then evokes the innate immune response. As an adaptor protein, STING plays a central role in controlling activation of the cGAS-STING pathway. Although transient activation of STING is essential to trigger the host defense during pathogen invasion, chronic STING activation has been shown to be associated with several autoinflammatory diseases. Here, we report that PPP6C negatively regulates the cGAS-STING pathway by removing STING phosphorylation, which is required for its activation. Dephosphorylation of STING by PPP6C helps prevent the sustained production of STING-dependent cytokines, which would otherwise lead to severe autoimmune disorders. This work provides additional mechanisms on the regulation of STING activity and might facilitate the development of novel therapeutics designed to prevent a variety of autoinflammatory disorders.


Assuntos
Herpesvirus Humano 1/genética , Imunidade Inata , Proteínas de Membrana/imunologia , Fosfoproteínas Fosfatases/imunologia , Vesiculovirus/genética , Animais , Chlorocebus aethiops , Regulação da Expressão Gênica , Células HEK293 , Herpesvirus Humano 1/fisiologia , Interações Hospedeiro-Patógeno , Humanos , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/imunologia , Fosfoproteínas Fosfatases/genética , Fosforilação , Células Vero , Vesiculovirus/fisiologia , Replicação Viral/genética , Replicação Viral/imunologia
5.
Protein Cell ; 11(8): 584-599, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32474700

RESUMO

The cyclic GMP-AMP (cGAMP) synthase (cGAS) plays a critical role in host defense by sensing cytosolic DNA derived from microbial pathogens or mis-located cellular DNA. Upon DNA binding, cGAS utilizes GTP and ATP as substrates to synthesize cGAMP, leading to MITA-mediated innate immune response. In this study, we identified the phosphatase PPP6C as a negative regulator of cGAS-mediated innate immune response. PPP6C is constitutively associated with cGAS in un-stimulated cells. DNA virus infection causes rapid disassociation of PPP6C from cGAS, resulting in phosphorylation of human cGAS S435 or mouse cGAS S420 in its catalytic pocket. Mutation of this serine residue of cGAS impairs its ability to synthesize cGAMP upon DNA virus infection. In vitro experiments indicate that S420-phosphorylated mcGAS has higher affinity to GTP and enzymatic activity. PPP6C-deficiency promotes innate immune response to DNA virus in various cells. Our findings suggest that PPP6C-mediated dephosphorylation of a catalytic pocket serine residue of cGAS impairs its substrate binding activity and innate immune response, which provides a mechanism for keeping the DNA sensor cGAS inactive in the absence of infection to avoid autoimmune response.


Assuntos
DNA Viral/imunologia , Imunidade Inata/imunologia , Nucleotidiltransferases/imunologia , Fosfoproteínas Fosfatases/imunologia , Animais , Sítios de Ligação , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Modelos Moleculares , Nucleotidiltransferases/deficiência , Nucleotidiltransferases/genética , Fosfoproteínas Fosfatases/deficiência , Fosforilação , Especificidade por Substrato , Células THP-1
6.
Sci Rep ; 10(1): 8323, 2020 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-32433485

RESUMO

Viral infections trigger host innate immune responses, characterized by the production of type-I interferons (IFN) including IFNß. IFNß induces cellular antiviral defense mechanisms and thereby contributes to pathogen clearance. Accumulating evidence suggests that mitochondria constitute a crucial platform for the induction of antiviral immunity. Here we demonstrate that the mitochondrial protein phosphoglycerate mutase family member 5 (PGAM5) is important for the antiviral cellular response. Following challenge of HeLa cells with the dsRNA-analog poly(I:C), PGAM5 oligomers and high levels of PGAM5 were found in mitochondrial aggregates. Using immunoprecipitation, a direct interaction of PGAM5 with the mitochondrial antiviral-signaling protein (MAVS) was demonstrated. In addition, PGAM5 deficient cells showed diminished expression of IFNß and IFNß target genes as compared to WT cells. Moreover, PGAM5 deficient mouse embryonic fibroblasts (MEFs) exhibited decreased phosphorylation levels of IRF3 and TBK1 when challenged with poly(I:C) intracellularly. Finally, PGAM5 deficient MEFs, upon infection with vesicular stomatitis virus (VSV), revealed diminished IFNß expression and increased VSV replication. Collectively, our study highlights PGAM5 as an important regulator for IFNß production mediated via the TBK1/IRF3 signaling pathway in response to viral infection.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Mitocondriais/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Vírus da Estomatite Vesicular Indiana/imunologia , Animais , Células Cultivadas , Fibroblastos/virologia , Células HeLa , Humanos , Fator Regulador 3 de Interferon/metabolismo , Interferon beta/metabolismo , Camundongos , Proteínas Mitocondriais/imunologia , Fosfoproteínas Fosfatases/imunologia , Poli I-C/imunologia , Proteínas Serina-Treonina Quinases/metabolismo , Infecções por Rhabdoviridae/imunologia , Transdução de Sinais , Replicação Viral/imunologia
7.
Biochem J ; 476(22): 3533-3548, 2019 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-31710352

RESUMO

Hypoxia in adipose tissue is regarded as a trigger that induces dysregulation of the secretory profile in adipocytes. Similarly, local dysregulation of adipocytokine secretion is an initial event in the deleterious effects of obesity on metabolism. We previously reported that CXCL13 is highly produced during adipogenesis, however little is known about the roles of CXCL13 in adipocytes. Here, we found that hypoxia, as modeled by 1% O2 or exposure to the hypoxia-mimetic reagent desferrioxamine (DFO) has strong inductive effects on the expression of CXCL13 and CXCR5, a CXCL13 receptor, in both undifferentiated and differentiated adipocytes and in organ-cultured white adipose tissue (WAT). CXCL13 was also highly expressed in WAT from high fat diet-fed mice. Hypoxic profile, typified by increased expression of interleukin-6 (IL-6) and plasminogen activator inhibitor-1 (PAI-1) and decreased expression of adiponectin, was significantly induced by CXCL13 treatment during adipogenic differentiation. Conversely, the treatment of adipocytes with a neutralizing-antibody against CXCL13 as well as CXCR5 knockdown by specific siRNA effectively inhibited DFO-induced inflammation. The phosphorylation of Akt2, a protective factor of adipose inflammation, was significantly inhibited by CXCL13 treatment during adipogenic differentiation. Mechanistically, CXCL13 induces the expression of PHLPP1, an Akt2 phosphatase, through focal adhesion kinase (FAK) signaling; and correspondingly we show that CXCL13 and DFO-induced IL-6 and PAI-1 expression was blocked by Phlpp1 knockdown. Furthermore, we revealed the functional binding sites of PPARγ2 and HIF1-α within the Cxcl13 promoter. Taken together, these results indicate that CXCL13 is an adipocytokine that facilitates hypoxia-induced inflammation in adipocytes through FAK-mediated induction of PHLPP1 in autocrine and/or paracrine manner.


Assuntos
Adipócitos/imunologia , Adipogenia , Adipocinas/imunologia , Quimiocina CXCL13/imunologia , Hipóxia/imunologia , Fosfoproteínas Fosfatases/imunologia , Células 3T3-L1 , Adipócitos/citologia , Adipocinas/genética , Adiponectina/genética , Adiponectina/imunologia , Animais , Quimiocina CXCL13/genética , Humanos , Hipóxia/genética , Hipóxia/fisiopatologia , Interleucina-6/genética , Interleucina-6/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , PPAR gama/genética , PPAR gama/imunologia , Fosfoproteínas Fosfatases/genética
8.
Front Immunol ; 10: 2169, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31608051

RESUMO

Ozone exposure causes irritation, airway hyperreactivity (AHR), inflammation of the airways, and destruction of alveoli (emphysema), the gas exchange area of the lung in human and mice. This review focuses on the acute disruption of the respiratory epithelial barrier in mice. A single high dose ozone exposure (1 ppm for 1 h) causes first a break of the bronchiolar epithelium within 2 h with leak of serum proteins in the broncho-alveolar space, disruption of epithelial tight junctions and cell death, which is followed at 6 h by ROS activation, AHR, myeloid cell recruitment, and remodeling. High ROS levels activate a novel PGAM5 phosphatase dependent cell-death pathway, called oxeiptosis. Bronchiolar cell wall damage and inflammation upon a single ozone exposure are reversible. However, chronic ozone exposure leads to progressive and irreversible loss of alveolar epithelial cells and alveoli with reduced gas exchange space known as emphysema. It is further associated with chronic inflammation and fibrosis of the lung, resembling other environmental pollutants and cigarette smoke in pathogenesis of asthma, and chronic obstructive pulmonary disease (COPD). Here, we review recent data on the mechanisms of ozone induced injury on the different cell types and pathways with a focus on the role of the IL-1 family cytokines and the related IL-33. The relation of chronic ozone exposure induced lung disease with asthma and COPD and the fact that ozone exacerbates asthma and COPD is emphasized.


Assuntos
Barreira Alveolocapilar/imunologia , Ozônio/toxicidade , Mucosa Respiratória/imunologia , Doença Aguda , Animais , Asma/induzido quimicamente , Asma/imunologia , Asma/patologia , Barreira Alveolocapilar/patologia , Fumar Cigarros/efeitos adversos , Fumar Cigarros/imunologia , Humanos , Camundongos , Fosfoproteínas Fosfatases/imunologia , Pneumonia/induzido quimicamente , Pneumonia/imunologia , Pneumonia/patologia , Doença Pulmonar Obstrutiva Crônica/induzido quimicamente , Doença Pulmonar Obstrutiva Crônica/imunologia , Doença Pulmonar Obstrutiva Crônica/patologia , Enfisema Pulmonar/induzido quimicamente , Enfisema Pulmonar/imunologia , Enfisema Pulmonar/patologia , Espécies Reativas de Oxigênio/imunologia , Mucosa Respiratória/patologia , Junções Íntimas/imunologia , Junções Íntimas/patologia
9.
Front Immunol ; 10: 2127, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31555304

RESUMO

Neutrophils are known to adopt dynamic and distinct functional phenotypes involved in the modulation of inflammation and immune homeostasis. However, inter-cellular signaling mechanisms that govern neutrophil polarization dynamics are not well understood. Employing a novel model of PHLPP deficient mice, we examined how neutrophils deficient in PHLPP may uniquely modulate immune defense and the host response during acute colitis. We found that PHLPP-/- mice were protected from dextran sodium sulfate (DSS)-induced septic colitis characterized by minimal body weight-loss, alleviated colon tissue destruction and reduced clinical symptoms. PHLPP-/- neutrophils have enhanced immune homeostasis as compared to WT neutrophils, reflected in enhanced migratory capacity toward chemoattractants, and reduced expression of inflammatory mediators due to elevated phosphorylation of AKT, STAT1, and ERK. Further, adoptive transfer of PHLPP deficient neutrophils to WT mice is sufficient to potently alleviate the severity of DSS-induced colitis. Our data reveal that PHLPP deficient neutrophils can be uniquely reprogrammed to a state conducive to host inflammation resolution. As a consequence, PHLPP-/- neutrophils can effectively transfer immune homeostasis in mice subjected to acute colitis. Our findings hold significant and novel insights into the mechanisms by which neutrophils can be effectively reprogrammed into a homeostatic state conducive for treating acute injuries such as septic colitis.


Assuntos
Colite/imunologia , Homeostase/imunologia , Neutrófilos/imunologia , Fosfoproteínas Fosfatases/imunologia , Animais , Colite/induzido quimicamente , Sulfato de Dextrana/imunologia , Sulfato de Dextrana/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infiltração de Neutrófilos/imunologia , Fosfoproteínas Fosfatases/deficiência
10.
PLoS One ; 14(8): e0221289, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31425567

RESUMO

The pathophysiological mechanisms of sepsis-induced cardiac dysfunction are largely unknown. The Toll-like receptor 4 (TLR4) is expressed in cardiac myocytes and is involved in bacterial endotoxin-mediated inflammatory disorders. TLR4 signaling leads to activation of the nuclear factor kappa B followed by increased expression of cytokines. Several protein phosphatases including PP2Cß, PP2A or PP1 are known to act as regulators of this signaling pathway. Here, we examined the role of PP5 for the inflammatory response to the bacterial endotoxin lipopolysaccharide in the heart using a transgenic mouse model with cardiac myocyte directed overexpression of PP5. In these transgenic mice, basal cardiac contractility was reduced, in vivo as well as in vitro, but LPS-induced cardiac dysfunction was less pronounced compared to wild type mice. Quantitative RT-PCR suggested an attenuated NF-κB signaling in the heart and cardiac expression of heat shock protein 25 (HSP25) was increased in PP5 transgenic mice. From our data we assume that PP5 increases stress tolerance of cardiac myocytes by downregulation of NF-κB signaling and upregulation of HSP25 expression.


Assuntos
Insuficiência Cardíaca/imunologia , Miócitos Cardíacos/imunologia , Proteínas Nucleares/imunologia , Fosfoproteínas Fosfatases/imunologia , Sepse/complicações , Receptor 4 Toll-Like/metabolismo , Animais , Modelos Animais de Doenças , Regulação para Baixo , Ecocardiografia , Feminino , Insuficiência Cardíaca/diagnóstico , Proteínas de Choque Térmico/metabolismo , Humanos , Preparação de Coração Isolado , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Camundongos Transgênicos , Chaperonas Moleculares/metabolismo , Contração Miocárdica/imunologia , Miócitos Cardíacos/metabolismo , NF-kappa B/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfoproteínas Fosfatases/genética , Fosfoproteínas Fosfatases/metabolismo , Sepse/imunologia , Sepse/microbiologia , Transdução de Sinais/imunologia , Regulação para Cima
11.
Proc Natl Acad Sci U S A ; 115(37): 9288-9293, 2018 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-30150402

RESUMO

Th17 cells favor glycolytic metabolism, and pyruvate dehydrogenase (PDH) is the key bifurcation enzyme, which in its active dephosphorylated form advances the oxidative phosphorylation from glycolytic pathway. The transcriptional factor, inducible cAMP early repressor/cAMP response element modulator (ICER/CREM), has been shown to be induced in Th17 cells and to be overexpressed in CD4+ T cells from the patients with systemic lupus erythematosus (SLE). We found that glycolysis and lactate production in in vitro Th17-polarized T cells was reduced and that the expression of pyruvate dehydrogenase phosphatase catalytic subunit 2 (PDP2), an enzyme that converts the inactive PDH to its active form, and PDH enzyme activity were increased in Th17 cells from ICER/CREM-deficient animals. ICER was found to bind to the Pdp2 promoter and suppress its expression. Furthermore, forced expression of PDP2 in CD4+ cells reduced the in vitro Th17 differentiation, whereas shRNA-based suppression of PDP2 expression increased in vitro Th17 differentiation and augmented experimental autoimmune encephalomyelitis. At the translational level, PDP2 expression was decreased in memory Th17 cells from patients with SLE and forced expression of PDP2 in CD4+ T cells from lupus-prone MRL/lpr mice and patients with SLE suppressed Th17 differentiation. These data demonstrate the direct control of energy production during Th17 differentiation in health and disease by the transcription factor ICER/CREM at the PDH metabolism bifurcation level.


Assuntos
Diferenciação Celular , Regulação Enzimológica da Expressão Gênica , Fosfoproteínas Fosfatases/biossíntese , Elementos de Resposta , Células Th17/enzimologia , Animais , Domínio Catalítico , Modulador de Elemento de Resposta do AMP Cíclico/genética , Modulador de Elemento de Resposta do AMP Cíclico/imunologia , Modulador de Elemento de Resposta do AMP Cíclico/metabolismo , Encefalomielite Autoimune Experimental/enzimologia , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Feminino , Humanos , Lúpus Eritematoso Sistêmico/enzimologia , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/patologia , Masculino , Camundongos , Camundongos Knockout , Fosfoproteínas Fosfatases/genética , Fosfoproteínas Fosfatases/imunologia , Células Th17/imunologia , Células Th17/patologia
12.
Biochem Biophys Res Commun ; 497(1): 80-86, 2018 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-29407174

RESUMO

Multiple sclerosis (MS) is a long-lasting autoimmune disease of the central nervous system. Currently, the etiology of MS is not known. Experimental autoimmune encephalomyelitis (EAE), has been recognized as the most widely used animal models to study the molecular mechanisms underlying MS and the efficacy of potential drugs for treatment of MS. In the present study, we found that Dl-3-n-butylphthalide (NBP), a neuroprotective drug in ischemic brain injury, prevented development of disease in experimental autoimmune encephalomyelitis (EAE) and significantly reduced inflammatory factors and necroptosis-associated genes, including PGAM5 in the spinal cord tissues. Similarly, silence of PGAM5 in spinal cord also ameliorated the disease severity in the mice with EAE. Moreover, re-expression of PGAM5 counteracted the protective effect of NBP on the pathogenesis of EAE. Importantly, we found that both NBP and silence of PGAM5 inhibited cellular necroptosis and inflammation in microglia induced by TNFα plus zVAD-fmk. Meanwhile, overexpression of PGAM5 reactivated cellular necroptosis and inflammation suppressed by NBP in vitro. Taken together, our findings provide evidence that NBP can attenuate the progression of EAE by suppressing PGAM5-induced necroptosis and inflammation in microglia and represents a new therapeutic strategy for treating autoimmune diseases.


Assuntos
Benzofuranos/administração & dosagem , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/imunologia , Microglia/imunologia , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/imunologia , Fosfoproteínas Fosfatases/imunologia , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Proteínas Reguladoras de Apoptose/imunologia , Relação Dose-Resposta a Droga , Encefalomielite Autoimune Experimental/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Esclerose Múltipla/patologia , Necrose/tratamento farmacológico , Necrose/imunologia , Necrose/patologia , Fármacos Neuroprotetores/administração & dosagem , Fosfoproteínas Fosfatases/antagonistas & inibidores , Resultado do Tratamento
13.
Mol Cell ; 68(2): 293-307.e5, 2017 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-29053956

RESUMO

Mitochondrial antiviral signaling platform protein (MAVS) acts as a central hub for RIG-I receptor proximal signal propagation. However, key components in the assembly of the MAVS mitochondrial platform that promote RIG-I mitochondrial localization and optimal activation are still largely undefined. Employing pooled RNAi and yeast two-hybrid screenings, we report that the mitochondrial adaptor protein tripartite motif (TRIM)14 provides a docking platform for the assembly of the mitochondrial signaling complex required for maximal activation of RIG-I-mediated signaling, consisting of WHIP and protein phosphatase PPP6C. Following viral infection, the ubiquitin-binding domain in WHIP bridges RIG-I with MAVS by binding to polyUb chains of RIG-I at lysine 164. The ATPase domain in WHIP contributes to stabilization of the RIG-I-dsRNA interaction. Moreover, phosphatase PPP6C is responsible for RIG-I dephosphorylation. Together, our findings define the WHIP-TRIM14-PPP6C mitochondrial signalosome required for RIG-I-mediated innate antiviral immunity.


Assuntos
Proteínas de Transporte/imunologia , Proteína DEAD-box 58/imunologia , Proteínas de Ligação a DNA/imunologia , Imunidade Inata , Mitocôndrias/imunologia , Proteínas Mitocondriais/imunologia , Complexos Multiproteicos/imunologia , Fosfoproteínas Fosfatases/imunologia , Transdução de Sinais/imunologia , ATPases Associadas a Diversas Atividades Celulares , Animais , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Chlorocebus aethiops , Proteína DEAD-box 58/genética , Proteínas de Ligação a DNA/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Mitocôndrias/genética , Proteínas Mitocondriais/genética , Complexos Multiproteicos/genética , Fosfoproteínas Fosfatases/genética , Receptores Imunológicos , Transdução de Sinais/genética , Proteínas com Motivo Tripartido , Células Vero , Viroses/genética , Viroses/imunologia , Vírus/genética , Vírus/imunologia
14.
J Cell Biol ; 216(12): 4107-4121, 2017 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-29055012

RESUMO

Salmonella enterica serovar Typhimurium exploits the host's type I interferon (IFN-I) response to induce receptor-interacting protein (RIP) kinase-mediated necroptosis in macrophages. However, the events that drive necroptosis execution downstream of IFN-I and RIP signaling remain elusive. In this study, we demonstrate that S Typhimurium infection causes IFN-I-mediated up-regulation of the mitochondrial phosphatase Pgam5 through RIP3. Pgam5 subsequently interacts with Nrf2, which sequesters Nrf2 in the cytosol, thereby repressing the transcription of Nrf2-dependent antioxidative genes. The impaired ability to respond to S Typhimurium-induced oxidative stress results in reactive oxygen species-mediated mitochondrial damage, energy depletion, transient induction of autophagy, and autophagic degradation of p62. Reduced p62 levels impair interaction of p62 with Keap1, which further decreases Nrf2 function and antioxidative responses to S Typhimurium infection, eventually leading to cell death. Collectively, we identify impaired Nrf2-dependent redox homeostasis as an important mechanism that promotes cell death downstream of IFN-I and RIP3 signaling in S Typhimurium-infected macrophages.


Assuntos
Apoptose/genética , Interferon Tipo I/imunologia , Macrófagos/imunologia , Fator 2 Relacionado a NF-E2/imunologia , Proteína Serina-Treonina Quinases de Interação com Receptores/imunologia , Salmonella typhimurium/fisiologia , Animais , Autofagia/genética , Células da Medula Óssea/imunologia , Células da Medula Óssea/microbiologia , Regulação da Expressão Gênica , Interferon Tipo I/genética , Proteína 1 Associada a ECH Semelhante a Kelch/genética , Proteína 1 Associada a ECH Semelhante a Kelch/imunologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/imunologia , Mitocôndrias/microbiologia , Fator 2 Relacionado a NF-E2/genética , Necrose/genética , Necrose/imunologia , Necrose/patologia , Estresse Oxidativo , Fosfoproteínas Fosfatases/genética , Fosfoproteínas Fosfatases/imunologia , Espécies Reativas de Oxigênio/imunologia , Espécies Reativas de Oxigênio/metabolismo , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/imunologia , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Proteína Sequestossoma-1/genética , Proteína Sequestossoma-1/imunologia
15.
Biochem Biophys Res Commun ; 486(2): 533-538, 2017 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-28322791

RESUMO

We have previously reported that bacterial endotoxin LPS attenuates expression of PHLPP, a ser/thr phosphatase, at both transcript and protein levels in different immune cells, however the underlying molecular mechanism is unknown and is of significant interest. Here, in line with the decreased transcript levels upon LPS treatment, we observed that LPS caused significant reduction in PHLPP promoter activity. We observed that SP1, a transcription factor frequently associated with inflammation, was recruited to the PHLPP promoter region. Ectopic expression of SP1 enhanced both transcript and protein levels of PHLPP while knockdown of SP1 or pharmacological inhibition of SP1 DNA binding by mithramycin reduced PHLPP expression. Moreover, over-expression of SP1 co-activators CBP/p300 augmented SP1 driven PHLPP promoter activity. Of note, LPS treatment depleted SP1 and CBP protein levels due to which recruitment of SP1 to PHLPP promoter was reduced. Further, we found that re-introduction of SP1 restored promoter activity and transcript levels of PHLPP in LPS stimulated cells. Collectively, our data revealed the molecular mechanism underlying the regulation of PHLPP expression during LPS induced macrophage inflammatory response.


Assuntos
Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Proteínas Nucleares/genética , Fosfoproteínas Fosfatases/genética , RNA Mensageiro/genética , Fator de Transcrição Sp1/genética , Animais , Linhagem Celular , Regulação da Expressão Gênica , Genes Reporter , Células HEK293 , Humanos , Luciferases/genética , Luciferases/metabolismo , Macrófagos/citologia , Macrófagos/imunologia , Camundongos , Monócitos/citologia , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/imunologia , Fosfoproteínas Fosfatases/antagonistas & inibidores , Fosfoproteínas Fosfatases/imunologia , Plicamicina/farmacologia , Regiões Promotoras Genéticas , RNA Mensageiro/imunologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Fator de Transcrição Sp1/imunologia , Transcrição Gênica , Fatores de Transcrição de p300-CBP/genética , Fatores de Transcrição de p300-CBP/imunologia
16.
PLoS Pathog ; 12(8): e1005811, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27494702

RESUMO

Plants recognize pathogen-associated molecular patterns (PAMPs) via cell surface-localized pattern recognition receptors (PRRs), leading to PRR-triggered immunity (PTI). The Arabidopsis cytoplasmic kinase BIK1 is a downstream substrate of several PRR complexes. How plant PTI is negatively regulated is not fully understood. Here, we identify the protein phosphatase PP2C38 as a negative regulator of BIK1 activity and BIK1-mediated immunity. PP2C38 dynamically associates with BIK1, as well as with the PRRs FLS2 and EFR, but not with the co-receptor BAK1. PP2C38 regulates PAMP-induced BIK1 phosphorylation and impairs the phosphorylation of the NADPH oxidase RBOHD by BIK1, leading to reduced oxidative burst and stomatal immunity. Upon PAMP perception, PP2C38 is phosphorylated on serine 77 and dissociates from the FLS2/EFR-BIK1 complexes, enabling full BIK1 activation. Together with our recent work on the control of BIK1 turnover, this study reveals another important regulatory mechanism of this central immune component.


Assuntos
Proteínas de Arabidopsis/imunologia , Arabidopsis/imunologia , Fosfoproteínas Fosfatases/imunologia , Imunidade Vegetal/fisiologia , Proteínas Serina-Treonina Quinases/imunologia , Arabidopsis/genética , Proteínas de Arabidopsis/genética , NADPH Oxidases/genética , NADPH Oxidases/imunologia , Fosfoproteínas Fosfatases/genética , Fosforilação/genética , Fosforilação/imunologia , Proteínas Quinases/genética , Proteínas Quinases/imunologia , Proteínas Serina-Treonina Quinases/genética
17.
J Immunol ; 196(12): 5056-63, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27183605

RESUMO

Leishmaniasis is an important parasitic disease found in the tropics and subtropics. Cutaneous and visceral leishmaniasis affect an estimated 1.5 million people worldwide. Despite its human health relevance, relatively little is known about the cell death pathways that control Leishmania replication in the host. Necroptosis is a recently identified form of cell death with potent antiviral effects. Receptor interacting protein kinase 1 (RIPK1) is a critical kinase that mediates necroptosis downstream of death receptors and TLRs. Heme, a product of hemoglobin catabolism during certain intracellular pathogen infections, is also a potent inducer of macrophage necroptosis. We found that human visceral leishmaniasis patients exhibit elevated serum levels of heme. Therefore, we examined the impact of heme and necroptosis on Leishmania replication. Indeed, heme potently inhibited Leishmania replication in bone marrow-derived macrophages. Moreover, we found that inhibition of RIPK1 kinase activity also enhanced parasite replication in the absence of heme. We further found that the mitochondrial phosphatase phosphoglycerate mutase family member 5 (PGAM5), a putative downstream effector of RIPK1, was also required for inhibition of Leishmania replication. In mouse infection, both PGAM5 and RIPK1 kinase activity are required for IL-1ß expression in response to Leishmania However, PGAM5, but not RIPK1 kinase activity, was directly responsible for Leishmania-induced IL-1ß secretion and NO production in bone marrow-derived macrophages. Collectively, these results revealed that RIPK1 and PGAM5 function independently to exert optimal control of Leishmania replication in the host.


Assuntos
Interações Hospedeiro-Parasita , Leishmania/crescimento & desenvolvimento , Leishmania/imunologia , Leishmaniose/parasitologia , Fosfoproteínas Fosfatases/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Animais , Morte Celular , Heme/análise , Heme/farmacologia , Humanos , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Interleucina-1beta/metabolismo , Leishmania/efeitos dos fármacos , Leishmaniose/sangue , Leishmaniose/imunologia , Leishmaniose/microbiologia , Leishmaniose Visceral/sangue , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/microbiologia , Macrófagos/fisiologia , Camundongos , Óxido Nítrico/biossíntese , Fosfoproteínas Fosfatases/genética , Fosfoproteínas Fosfatases/imunologia , Proteína Serina-Treonina Quinases de Interação com Receptores/antagonistas & inibidores
18.
Int Immunopharmacol ; 34: 229-234, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26971226

RESUMO

PH domain leucine-rich repeats protein phosphatase 1(PHLPP1) belongs to a novel family of Ser/Thr protein phosphatases: PHLPP serves as tumor suppressor in several cancers. However, little knowledge about the expression of PHLPP1 in human glioma tumor tissue and its role in inflammation response in glioma cells was known. Glioma samples were obtained from a total of 37 patients including 16 males and 21 females with surgical removal of the brain tumor. PHLPP1 protein and inflammatory cytokines were measured by Western blot analysis and immunohistochemistry while mRNA was determined by RT-PCR. The levels of inflammatory cytokines including TNF-α, IL-17, IL-1ß in U251 glioma cells were evaluated by siRNA PHLPP1 and PHLPP1 addition. The loss of PHLPP1 expression occurs at high frequency in human gliomas. The highest mean values of PHLPP1 mRNA and protein were found in non-glioma brain tissues whereas the lowest mean values were found in those in glioblastoma with an increase of TNF-α, IL-17, IL-1ß (p<0.05). PHLPP1 expression in human glioma was associated negatively with the severity of the tumor and inflammatory cytokines. siRNA PHLPP1 could increase the levels of inflammatory cytokines in U251 glioma cells while PHLPP1 addition could inhibit significantly inflammatory cytokines. We concluded that PHLPP1 played a suppression role in inflammatory response of glioma. The present study indicated that PHLPP1 could be used as a predictor for the prediction of the patients or as a therapeutic target for the treatment of human glioma.


Assuntos
Neoplasias Encefálicas/imunologia , Glioma/imunologia , Proteínas Nucleares/imunologia , Fosfoproteínas Fosfatases/imunologia , Proteínas Supressoras de Tumor/metabolismo , Neoplasias Encefálicas/patologia , Carcinogênese , Linhagem Celular , Citocinas/genética , Citocinas/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Glioma/patologia , Humanos , Mediadores da Inflamação/metabolismo , Masculino , Estadiamento de Neoplasias , Proteínas Nucleares/genética , Fosfoproteínas Fosfatases/genética , RNA Interferente Pequeno/genética , Proteínas Supressoras de Tumor/genética
19.
J Innate Immun ; 8(2): 171-84, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26457672

RESUMO

Endotoxin tolerance protects the host by limiting excessive 'cytokine storm' during sepsis, but compromises the ability to counteract infections in septic shock survivors. It reprograms Toll-like receptor (TLR) 4 responses by attenuating the expression of proinflammatory cytokines without suppressing anti-inflammatory and antimicrobial mediators, but the mechanisms of reprogramming remain unclear. In this study, we demonstrate that the induction of endotoxin tolerance in human monocytes, THP-1 and MonoMac-6 cells inhibited lipopolysaccharide (LPS)-mediated phosphorylation of Lyn, c-Src and their recruitment to TLR4, but increased total protein phosphatase (PP) activity and the expression of protein tyrosine phosphatase (PTP) 1B, PP2A, PTP nonreceptor type (PTPN) 22 and mitogen-activated protein kinase phosphatase (MKP)-1. Chemical PP inhibitors, okadaic acid, dephostatin and cantharidic acid markedly decreased or completely abolished LPS tolerance, indicating the importance of phosphatases in endotoxin tolerization. Overexpression of PTPN22 decreased LPS-mediated nuclear factor (NF)-x03BA;B activation, p38 phosphorylation and CXCL8 gene expression, while PTPN22 ablation upregulated LPS-induced p65 NF-x03BA;B and p38 phosphorylation and the expression of TNF-α and pro-IL-1ß mRNA, indicating PTPN22 as an inhibitor of TLR4 signaling. Thus, LPS tolerance interferes with TLR4 signaling by inhibiting Lyn and c-Src phosphorylation and their recruitment to TLR4, while increasing the phosphatase activity and expression of PP2A, PTPN22, PTP1B and MKP1.


Assuntos
Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Tolerância Imunológica/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Monócitos/imunologia , Fosfoproteínas Fosfatases/imunologia , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/imunologia , Quinases da Família src/imunologia , Proteína Tirosina Quinase CSK , Linhagem Celular Tumoral , Regulação Enzimológica da Expressão Gênica/imunologia , Humanos , Tolerância Imunológica/genética , Lipopolissacarídeos/imunologia , Fosfoproteínas Fosfatases/genética , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Receptor 4 Toll-Like/genética , Quinases da Família src/genética
20.
J Immunol ; 195(8): 3849-57, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26363053

RESUMO

The effective recognition of viral infection and subsequent type I IFN production is essential for the host antiviral innate immune responses. The phosphorylation and activation of kinase TANK-binding kinase 1 (TBK1) plays crucial roles in the production of type I IFN mediated by TLR and retinoic acid-inducible gene I-like receptors. Type I IFN expression must be tightly regulated to prevent the development of immunopathological disorders. However, how the activated TBK1 is negatively regulated by phosphatases remains poorly understood. In this study, we identified a previously unknown role of protein phosphatase (PP)4 by acting as a TBK1 phosphatase. PP4 expression was upregulated in macrophages infected with RNA virus, vesicular stomatitis virus, and Sendai virus in vitro and in vivo. Knockdown of PP4C, the catalytic subunit of PP4, significantly increased type I IFN production in macrophages and dentritic cells triggered by TLR3/4 ligands, vesicular stomatitis virus, and Sendai virus, and thus inhibited virus replication. Similar results were also found in peritoneal macrophages with PP4C silencing in vivo and i.p. infection of RNA virus. Accordingly, ectopic expression of PP4C inhibited virus-induced type I IFN production and promoted virus replication. However, overexpression of a phosphatase-dead PP4C mutant abolished the inhibitory effects of wild-type PP4C on type I IFN production. Mechanistically, PP4 directly bound TBK1 upon virus infection, then dephosphorylated TBK1 at Ser(172) and inhibited TBK1 activation, and subsequently restrained IFN regulatory factor 3 activation, resulting in suppressed production of type I IFN and IFN-stimulated genes. Thus, serine/threonine phosphatase PP4 functions as a novel feedback negative regulator of RNA virus-triggered innate immunity.


Assuntos
Regulação da Expressão Gênica/imunologia , Imunidade Inata , Interferon Tipo I/imunologia , Fosfoproteínas Fosfatases/imunologia , Infecções por Respirovirus/imunologia , Infecções por Rhabdoviridae/imunologia , Vírus Sendai/fisiologia , Vesiculovirus/fisiologia , Replicação Viral/imunologia , Animais , Células Dendríticas/imunologia , Células Dendríticas/patologia , Macrófagos/imunologia , Macrófagos/patologia , Camundongos , Fosforilação/imunologia , Proteínas Serina-Treonina Quinases/imunologia , Infecções por Respirovirus/patologia , Infecções por Rhabdoviridae/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA