Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 150
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 119(1)2022 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-34930823

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas , Coxiella burnetii , Fosfoproteínas Fosfatasas , Fiebre Q , Transducción de Señal , Factores de Virulencia , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Chlorocebus aethiops , Coxiella burnetii/genética , Coxiella burnetii/inmunología , Coxiella burnetii/patogenicidad , Células HEK293 , Células HeLa , Humanos , FN-kappa B/genética , FN-kappa B/inmunología , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/inmunología , Fiebre Q/genética , Fiebre Q/inmunología , Transducción de Señal/genética , Transducción de Señal/inmunología , Células Vero , Factores de Virulencia/genética , Factores de Virulencia/inmunología
2.
J Neuroinflammation ; 18(1): 145, 2021 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-34183017

RESUMEN

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.


Asunto(s)
Sistema Nervioso Central/patología , Encefalomielitis Autoinmune Experimental , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Interferón gamma/metabolismo , Microglía/inmunología , Fosfoproteínas Fosfatasas/inmunología , Receptores de Hidrocarburo de Aril/metabolismo , Animales , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/fisiopatología , Citocinas , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Técnicas de Inactivación de Genes , Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Esclerosis Múltiple/inmunología , Glicoproteína Mielina-Oligodendrócito/inmunología , Células Mieloides/inmunología , Células Mieloides/metabolismo , Fragmentos de Péptidos/inmunología , Fosfoproteínas Fosfatasas/metabolismo , Transducción de Señal , Bazo/patología
3.
Clin Exp Immunol ; 203(1): 41-46, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32979862

RESUMEN

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.


Asunto(s)
Autoanticuerpos/sangre , Diabetes Mellitus Tipo 1/sangre , Islotes Pancreáticos/metabolismo , Adolescente , Autoanticuerpos/inmunología , Niño , Preescolar , Diabetes Mellitus Tipo 1/inmunología , Inglaterra , Femenino , Glutamato Descarboxilasa/inmunología , Glutamato Descarboxilasa/metabolismo , Humanos , Islotes Pancreáticos/inmunología , Lituania , Masculino , Fosfoproteínas Fosfatasas/inmunología , Fosfoproteínas Fosfatasas/metabolismo , Transportador 8 de Zinc/inmunología , Transportador 8 de Zinc/metabolismo
4.
mBio ; 11(4)2020 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-32753499

RESUMEN

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.


Asunto(s)
Herpesvirus Humano 1/genética , Inmunidad Innata , Proteínas de la Membrana/inmunología , Fosfoproteínas Fosfatasas/inmunología , Vesiculovirus/genética , Animales , Chlorocebus aethiops , Regulación de la Expresión Génica , Células HEK293 , Herpesvirus Humano 1/fisiología , Interacciones Huésped-Patógeno , Humanos , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/inmunología , Fosfoproteínas Fosfatasas/genética , Fosforilación , Células Vero , Vesiculovirus/fisiología , Replicación Viral/genética , Replicación Viral/inmunología
5.
Protein Cell ; 11(8): 584-599, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32474700

RESUMEN

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.


Asunto(s)
ADN Viral/inmunología , Inmunidad Innata/inmunología , Nucleotidiltransferasas/inmunología , Fosfoproteínas Fosfatasas/inmunología , Animales , Sitios de Unión , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Nucleotidiltransferasas/deficiencia , Nucleotidiltransferasas/genética , Fosfoproteínas Fosfatasas/deficiencia , Fosforilación , Especificidad por Sustrato , Células THP-1
6.
Sci Rep ; 10(1): 8323, 2020 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-32433485

RESUMEN

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.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Mitocondriales/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Virus de la Estomatitis Vesicular Indiana/inmunología , Animales , Células Cultivadas , Fibroblastos/virología , Células HeLa , Humanos , Factor 3 Regulador del Interferón/metabolismo , Interferón beta/metabolismo , Ratones , Proteínas Mitocondriales/inmunología , Fosfoproteínas Fosfatasas/inmunología , Poli I-C/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Infecciones por Rhabdoviridae/inmunología , Transducción de Señal , Replicación Viral/inmunología
7.
Biochem J ; 476(22): 3533-3548, 2019 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-31710352

RESUMEN

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.


Asunto(s)
Adipocitos/inmunología , Adipogénesis , Adipoquinas/inmunología , Quimiocina CXCL13/inmunología , Hipoxia/inmunología , Fosfoproteínas Fosfatasas/inmunología , Células 3T3-L1 , Adipocitos/citología , Adipoquinas/genética , Adiponectina/genética , Adiponectina/inmunología , Animales , Quimiocina CXCL13/genética , Humanos , Hipoxia/genética , Hipoxia/fisiopatología , Interleucina-6/genética , Interleucina-6/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , PPAR gamma/genética , PPAR gamma/inmunología , Fosfoproteínas Fosfatasas/genética
8.
Front Immunol ; 10: 2169, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31608051

RESUMEN

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.


Asunto(s)
Barrera Alveolocapilar/inmunología , Ozono/toxicidad , Mucosa Respiratoria/inmunología , Enfermedad Aguda , Animales , Asma/inducido químicamente , Asma/inmunología , Asma/patología , Barrera Alveolocapilar/patología , Fumar Cigarrillos/efectos adversos , Fumar Cigarrillos/inmunología , Humanos , Ratones , Fosfoproteínas Fosfatasas/inmunología , Neumonía/inducido químicamente , Neumonía/inmunología , Neumonía/patología , Enfermedad Pulmonar Obstructiva Crónica/inducido químicamente , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Enfermedad Pulmonar Obstructiva Crónica/patología , Enfisema Pulmonar/inducido químicamente , Enfisema Pulmonar/inmunología , Enfisema Pulmonar/patología , Especies Reactivas de Oxígeno/inmunología , Mucosa Respiratoria/patología , Uniones Estrechas/inmunología , Uniones Estrechas/patología
9.
Front Immunol ; 10: 2127, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31555304

RESUMEN

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.


Asunto(s)
Colitis/inmunología , Homeostasis/inmunología , Neutrófilos/inmunología , Fosfoproteínas Fosfatasas/inmunología , Animales , Colitis/inducido químicamente , Sulfato de Dextran/inmunología , Sulfato de Dextran/toxicidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila/inmunología , Fosfoproteínas Fosfatasas/deficiencia
10.
PLoS One ; 14(8): e0221289, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31425567

RESUMEN

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.


Asunto(s)
Insuficiencia Cardíaca/inmunología , Miocitos Cardíacos/inmunología , Proteínas Nucleares/inmunología , Fosfoproteínas Fosfatasas/inmunología , Sepsis/complicaciones , Receptor Toll-Like 4/metabolismo , Animales , Modelos Animales de Enfermedad , Regulación hacia Abajo , Ecocardiografía , Femenino , Insuficiencia Cardíaca/diagnóstico , Proteínas de Choque Térmico/metabolismo , Humanos , Preparación de Corazón Aislado , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Transgénicos , Chaperonas Moleculares/metabolismo , Contracción Miocárdica/inmunología , Miocitos Cardíacos/metabolismo , FN-kappa B/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/metabolismo , Sepsis/inmunología , Sepsis/microbiología , Transducción de Señal/inmunología , Regulación hacia Arriba
11.
Proc Natl Acad Sci U S A ; 115(37): 9288-9293, 2018 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-30150402

RESUMEN

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.


Asunto(s)
Diferenciación Celular , Regulación Enzimológica de la Expresión Génica , Fosfoproteínas Fosfatasas/biosíntesis , Elementos de Respuesta , Células Th17/enzimología , Animales , Dominio Catalítico , Modulador del Elemento de Respuesta al AMP Cíclico/genética , Modulador del Elemento de Respuesta al AMP Cíclico/inmunología , Modulador del Elemento de Respuesta al AMP Cíclico/metabolismo , Encefalomielitis Autoinmune Experimental/enzimología , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Humanos , Lupus Eritematoso Sistémico/enzimología , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Masculino , Ratones , Ratones Noqueados , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/inmunología , Células Th17/inmunología , Células Th17/patología
12.
Biochem Biophys Res Commun ; 497(1): 80-86, 2018 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-29407174

RESUMEN

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.


Asunto(s)
Benzofuranos/administración & dosificación , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/inmunología , Microglía/inmunología , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/inmunología , Fosfoproteínas Fosfatasas/inmunología , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Proteínas Reguladoras de la Apoptosis/inmunología , Relación Dosis-Respuesta a Droga , Encefalomielitis Autoinmune Experimental/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Esclerosis Múltiple/patología , Necrosis/tratamiento farmacológico , Necrosis/inmunología , Necrosis/patología , Fármacos Neuroprotectores/administración & dosificación , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Resultado del Tratamiento
13.
Mol Cell ; 68(2): 293-307.e5, 2017 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-29053956

RESUMEN

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.


Asunto(s)
Proteínas Portadoras/inmunología , Proteína 58 DEAD Box/inmunología , Proteínas de Unión al ADN/inmunología , Inmunidad Innata , Mitocondrias/inmunología , Proteínas Mitocondriales/inmunología , Complejos Multiproteicos/inmunología , Fosfoproteínas Fosfatasas/inmunología , Transducción de Señal/inmunología , ATPasas Asociadas con Actividades Celulares Diversas , Animales , Proteínas Portadoras/genética , Línea Celular Tumoral , Chlorocebus aethiops , Proteína 58 DEAD Box/genética , Proteínas de Unión al ADN/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular , Mitocondrias/genética , Proteínas Mitocondriales/genética , Complejos Multiproteicos/genética , Fosfoproteínas Fosfatasas/genética , Receptores Inmunológicos , Transducción de Señal/genética , Proteínas de Motivos Tripartitos , Células Vero , Virosis/genética , Virosis/inmunología , Virus/genética , Virus/inmunología
14.
J Cell Biol ; 216(12): 4107-4121, 2017 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-29055012

RESUMEN

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.


Asunto(s)
Apoptosis/genética , Interferón Tipo I/inmunología , Macrófagos/inmunología , Factor 2 Relacionado con NF-E2/inmunología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/inmunología , Salmonella typhimurium/fisiología , Animales , Autofagia/genética , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/microbiología , Regulación de la Expresión Génica , Interferón Tipo I/genética , Proteína 1 Asociada A ECH Tipo Kelch/genética , Proteína 1 Asociada A ECH Tipo Kelch/inmunología , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/inmunología , Mitocondrias/microbiología , Factor 2 Relacionado con NF-E2/genética , Necrosis/genética , Necrosis/inmunología , Necrosis/patología , Estrés Oxidativo , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/inmunología , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/inmunología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Sequestosoma-1/genética , Proteína Sequestosoma-1/inmunología
15.
Biochem Biophys Res Commun ; 486(2): 533-538, 2017 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-28322791

RESUMEN

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.


Asunto(s)
Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Proteínas Nucleares/genética , Fosfoproteínas Fosfatasas/genética , ARN Mensajero/genética , Factor de Transcripción Sp1/genética , Animales , Línea Celular , Regulación de la Expresión Génica , Genes Reporteros , Células HEK293 , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Macrófagos/citología , Macrófagos/inmunología , Ratones , Monocitos/citología , Monocitos/efectos de los fármacos , Monocitos/inmunología , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/inmunología , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Fosfoproteínas Fosfatasas/inmunología , Plicamicina/farmacología , Regiones Promotoras Genéticas , ARN Mensajero/inmunología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Factor de Transcripción Sp1/inmunología , Transcripción Genética , Factores de Transcripción p300-CBP/genética , Factores de Transcripción p300-CBP/inmunología
16.
PLoS Pathog ; 12(8): e1005811, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27494702

RESUMEN

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.


Asunto(s)
Proteínas de Arabidopsis/inmunología , Arabidopsis/inmunología , Fosfoproteínas Fosfatasas/inmunología , Inmunidad de la Planta/fisiología , Proteínas Serina-Treonina Quinasas/inmunología , Arabidopsis/genética , Proteínas de Arabidopsis/genética , NADPH Oxidasas/genética , NADPH Oxidasas/inmunología , Fosfoproteínas Fosfatasas/genética , Fosforilación/genética , Fosforilación/inmunología , Proteínas Quinasas/genética , Proteínas Quinasas/inmunología , Proteínas Serina-Treonina Quinasas/genética
17.
J Immunol ; 196(12): 5056-63, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27183605

RESUMEN

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.


Asunto(s)
Interacciones Huésped-Parásitos , Leishmania/crecimiento & desarrollo , Leishmania/inmunología , Leishmaniasis/parasitología , Fosfoproteínas Fosfatasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Animales , Muerte Celular , Hemo/análisis , Hemo/farmacología , Humanos , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Leishmania/efectos de los fármacos , Leishmaniasis/sangre , Leishmaniasis/inmunología , Leishmaniasis/microbiología , Leishmaniasis Visceral/sangre , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/microbiología , Macrófagos/fisiología , Ratones , Óxido Nítrico/biosíntesis , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/inmunología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores
18.
Int Immunopharmacol ; 34: 229-234, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26971226

RESUMEN

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.


Asunto(s)
Neoplasias Encefálicas/inmunología , Glioma/inmunología , Proteínas Nucleares/inmunología , Fosfoproteínas Fosfatasas/inmunología , Proteínas Supresoras de Tumor/metabolismo , Neoplasias Encefálicas/patología , Carcinogénesis , Línea Celular , Citocinas/genética , Citocinas/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Glioma/patología , Humanos , Mediadores de Inflamación/metabolismo , Masculino , Estadificación de Neoplasias , Proteínas Nucleares/genética , Fosfoproteínas Fosfatasas/genética , ARN Interferente Pequeño/genética , Proteínas Supresoras de Tumor/genética
19.
J Innate Immun ; 8(2): 171-84, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26457672

RESUMEN

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.


Asunto(s)
Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Tolerancia Inmunológica/efectos de los fármacos , Lipopolisacáridos/farmacología , Monocitos/inmunología , Fosfoproteínas Fosfatasas/inmunología , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 4/inmunología , Familia-src Quinasas/inmunología , Proteína Tirosina Quinasa CSK , Línea Celular Tumoral , Regulación Enzimológica de la Expresión Génica/inmunología , Humanos , Tolerancia Inmunológica/genética , Lipopolisacáridos/inmunología , Fosfoproteínas Fosfatasas/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Receptor Toll-Like 4/genética , Familia-src Quinasas/genética
20.
J Immunol ; 195(8): 3849-57, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26363053

RESUMEN

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.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Inmunidad Innata , Interferón Tipo I/inmunología , Fosfoproteínas Fosfatasas/inmunología , Infecciones por Respirovirus/inmunología , Infecciones por Rhabdoviridae/inmunología , Virus Sendai/fisiología , Vesiculovirus/fisiología , Replicación Viral/inmunología , Animales , Células Dendríticas/inmunología , Células Dendríticas/patología , Macrófagos/inmunología , Macrófagos/patología , Ratones , Fosforilación/inmunología , Proteínas Serina-Treonina Quinasas/inmunología , Infecciones por Respirovirus/patología , Infecciones por Rhabdoviridae/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA