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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 50(6): 1412-1424.e6, 2019 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-31076360

RESUMEN

Assembly of inflammasomes after infection or injury leads to the release of interleukin-1ß (IL-1ß) and to pyroptosis. After inflammasome activation, cells either pyroptose or enter a hyperactivated state defined by IL-1ß secretion without cell death, but what controls these different outcomes is unknown. Here, we show that removal of the Toll-IL-1R protein SARM from macrophages uncouples inflammasome-dependent cytokine release and pyroptosis, whereby cells displayed increased IL-1ß production but reduced pyroptosis. Correspondingly, increasing SARM in cells caused less IL-1ß release and more pyroptosis. SARM suppressed IL-1ß by directly restraining the NLRP3 inflammasome and, hence, caspase-1 activation. Consistent with a role for SARM in pyroptosis, Sarm1-/- mice were protected from lipopolysaccharide (LPS)-stimulated sepsis. Pyroptosis-inducing, but not hyperactivating, NLRP3 stimulants caused SARM-dependent mitochondrial depolarization. Thus, SARM-dependent mitochondrial depolarization distinguishes NLRP3 activators that cause pyroptosis from those that do not, and SARM modulation represents a cell-intrinsic mechanism to regulate cell fate after inflammasome activation.


Asunto(s)
Proteínas del Dominio Armadillo/metabolismo , Citocinas/metabolismo , Proteínas del Citoesqueleto/metabolismo , Inflamasomas/metabolismo , Animales , Proteínas del Dominio Armadillo/genética , Biomarcadores , Supervivencia Celular , Proteínas del Citoesqueleto/genética , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Mitocondrias/genética , Mitocondrias/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Unión Proteica , Piroptosis , Transducción de Señal
2.
Cell ; 147(2): 259-61, 2011 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-22000004

RESUMEN

The innate immune sensor RIG-I responds to infection by binding to viral double-stranded RNA (dsRNA). In this issue of Cell, Kowalinski et al. (2011) and Luo et al. (2011) reveal the structure of RIG-I, and in combination with functional analyses, they show how RIG-I recognizes viral RNA to initiate signaling and a type I interferon response.

3.
Mol Cell ; 71(5): 745-760.e5, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-30193098

RESUMEN

DNA damage can be sensed as a danger-associated molecular pattern by the innate immune system. Here we find that keratinocytes and other human cells mount an innate immune response within hours of etoposide-induced DNA damage, which involves the DNA sensing adaptor STING but is independent of the cytosolic DNA receptor cGAS. This non-canonical activation of STING is mediated by the DNA binding protein IFI16, together with the DNA damage response factors ATM and PARP-1, resulting in the assembly of an alternative STING signaling complex that includes the tumor suppressor p53 and the E3 ubiquitin ligase TRAF6. TRAF6 catalyzes the formation of K63-linked ubiquitin chains on STING, leading to the activation of the transcription factor NF-κB and the induction of an alternative STING-dependent gene expression program. We propose that STING acts as a signaling hub that coordinates a transcriptional response depending on its mode of activation.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/genética , Núcleo Celular/genética , Daño del ADN/genética , Proteínas de la Membrana/genética , FN-kappa B/genética , Proteínas Nucleares/genética , Fosfoproteínas/genética , Transducción de Señal/genética , Línea Celular , Citosol/metabolismo , ADN/genética , Células HEK293 , Humanos , Inmunidad Innata/genética , Queratinocitos/fisiología , Poli(ADP-Ribosa) Polimerasa-1/genética , Proteína p53 Supresora de Tumor/genética , Ubiquitina/genética , Ubiquitina-Proteína Ligasas/genética
4.
J Biol Chem ; 300(2): 105620, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38176648

RESUMEN

Sterile alpha and HEAT/armadillo motif-containing protein (SARM1) was recently described as a NAD+-consuming enzyme and has previously been shown to regulate immune responses in macrophages. Neuronal SARM1 is known to contribute to axon degeneration due to its NADase activity. However, how SARM1 affects macrophage metabolism has not been explored. Here, we show that macrophages from Sarm1-/- mice display elevated NAD+ concentrations and lower cyclic ADP-ribose, a known product of SARM1-dependent NAD+ catabolism. Further, SARM1-deficient macrophages showed an increase in the reserve capacity of oxidative phosphorylation and glycolysis compared to WT cells. Stimulation of macrophages to a proinflammatory state by lipopolysaccharide (LPS) revealed that SARM1 restricts the ability of macrophages to upregulate glycolysis and limits the expression of the proinflammatory gene interleukin (Il) 1b, but boosts expression of anti-inflammatory Il10. In contrast, we show macrophages lacking SARM1 induced to an anti-inflammatory state by IL-4 stimulation display increased oxidative phosphorylation and glycolysis, and reduced expression of the anti-inflammatory gene, Fizz1. Overall, these data show that SARM1 fine-tunes immune gene transcription in macrophages via consumption of NAD+ and altered macrophage metabolism.


Asunto(s)
Proteínas del Dominio Armadillo , Proteínas del Citoesqueleto , Neuronas , Animales , Ratones , Proteínas del Dominio Armadillo/genética , Proteínas del Dominio Armadillo/metabolismo , Axones/metabolismo , ADP-Ribosa Cíclica/metabolismo , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , NAD/metabolismo , Neuronas/metabolismo
5.
Nat Immunol ; 19(1): 4-5, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29242544
6.
Immunity ; 44(3): 597-608, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26944200

RESUMEN

The cationic polysaccharide chitosan is an attractive candidate adjuvant capable of driving potent cell-mediated immunity, but the mechanism by which it acts is not clear. We show that chitosan promotes dendritic cell maturation by inducing type I interferons (IFNs) and enhances antigen-specific T helper 1 (Th1) responses in a type I IFN receptor-dependent manner. The induction of type I IFNs, IFN-stimulated genes and dendritic cell maturation by chitosan required the cytoplasmic DNA sensor cGAS and STING, implicating this pathway in dendritic cell activation. Additionally, this process was dependent on mitochondrial reactive oxygen species and the presence of cytoplasmic DNA. Chitosan-mediated enhancement of antigen specific Th1 and immunoglobulin G2c responses following vaccination was dependent on both cGAS and STING. These findings demonstrate that a cationic polymer can engage the STING-cGAS pathway to trigger innate and adaptive immune responses.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Quitosano/administración & dosificación , Células Dendríticas/fisiología , Proteínas de la Membrana/metabolismo , Mitocondrias/metabolismo , Nucleotidiltransferasas/metabolismo , Células TH1/inmunología , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Movimiento Celular , Células Cultivadas , ADN/metabolismo , Células Dendríticas/efectos de los fármacos , Femenino , Humanos , Inmunidad Celular/efectos de los fármacos , Inmunidad Celular/genética , Inmunoglobulina G/metabolismo , Interferón Tipo I/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Nucleotidiltransferasas/genética , Especies Reactivas de Oxígeno/metabolismo , Vacunas/administración & dosificación
7.
J Gen Virol ; 105(8)2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39167082

RESUMEN

Molluscum contagiosum virus (MCV) is a human-specific poxvirus that causes a highly common but mild infection characterized by distinctive and persistent papular skin lesions. These lesions can persist for long periods without an effective clearance response from the host. MCV, like all poxviruses, encodes multiple known immunosuppressive proteins which target innate immune signalling pathways involved in viral nucleic acid sensing, interferon production and inflammation which should trigger antiviral immunity leading to clearance. Two major families of transcription factors responsible for driving the immune response to viruses are the NF-κB and the interferon regulatory factor (IRF) families. While NF-κB broadly drives pro-inflammatory gene expression and IRFs chiefly drive interferon induction, both collaborate in transactivating many of the same genes in a concerted immune response to viral infection. Here, we report that the MCV protein MC089 specifically inhibits IRF activation from both DNA- and RNA-sensing pathways, making it the first characterized MCV inhibitor to selectively target IRF activation to date. MC089 interacts with proteins required for IRF activation, namely IKKε, TBKBP1 and NAP1. Additionally, MC089 targets RNA sensing by associating with the RNA-sensing adaptor protein mitochondrial antiviral-signalling protein on mitochondria. MC089 displays specificity in its inhibition of IRF3 activation by suppressing immunostimulatory nucleic acid-induced serine 396 phosphorylation without affecting the phosphorylation of serine 386. The selective interaction of MC089 with IRF-regulatory proteins and site-specific inhibition of IRF3 phosphorylation may offer a tool to provide novel insights into the biology of IRF3 regulation.


Asunto(s)
Factor 3 Regulador del Interferón , Virus del Molusco Contagioso , Proteínas Virales , Humanos , Factor 3 Regulador del Interferón/metabolismo , Factor 3 Regulador del Interferón/genética , Virus del Molusco Contagioso/inmunología , Virus del Molusco Contagioso/genética , Proteínas Virales/metabolismo , Proteínas Virales/genética , Proteínas Virales/inmunología , Transducción de Señal , Inmunidad Innata , Células HEK293 , Interacciones Huésped-Patógeno/inmunología
8.
J Virol ; 97(3): e0010823, 2023 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-36916940

RESUMEN

Molluscum contagiosum virus (MCV) is a human-adapted poxvirus that causes a common and persistent yet mild infection characterized by distinct, contagious, papular skin lesions. These lesions are notable for having little or no inflammation associated with them and can persist for long periods without an effective clearance response from the host. Like all poxviruses, MCV encodes potent immunosuppressive proteins that perturb innate immune pathways involved in virus sensing, the interferon response, and inflammation, which collectively orchestrate antiviral immunity and clearance, with several of these pathways converging at common signaling nodes. One such node is the regulator of canonical nuclear factor kappa B (NF-κB) activation, NF-κB essential modulator (NEMO). Here, we report that the MCV protein MC008 specifically inhibits NF-κB through its interaction with NEMO, disrupting its early ubiquitin-mediated activation and subsequent downstream signaling. MC008 is the third NEMO-targeting inhibitor to be described in MCV to date, with each inhibiting NEMO activation in distinct ways, highlighting strong selective pressure to evolve multiple ways of disabling this key signaling protein. IMPORTANCE Inflammation lies at the heart of most human diseases. Understanding the pathways that drive this response is the key to new anti-inflammatory therapies. Viruses evolve to target inflammation; thus, understanding how they do this reveals how inflammation is controlled and, potentially, how to disable it when it drives disease. Molluscum contagiosum virus (MCV) has specifically evolved to infect humans and displays an unprecedented ability to suppress inflammation in our tissue. We have identified a novel inhibitor of human innate signaling from MCV, MC008, which targets NEMO, a core regulator of proinflammatory signaling. Furthermore, MC008 appears to inhibit early ubiquitination, thus interrupting later events in NEMO activation, thereby validating current models of IκB kinase (IKK) complex regulation.


Asunto(s)
Virus del Molusco Contagioso , FN-kappa B , Humanos , FN-kappa B/metabolismo , Virus del Molusco Contagioso/metabolismo , Proteínas Virales/metabolismo , Transducción de Señal , Ubiquitinación , Quinasa I-kappa B/metabolismo
9.
Am J Respir Crit Care Med ; 205(5): 550-562, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-34985402

RESUMEN

Rationale: The Toll-like receptor 3 Leu412Phe (TLR3 L412F) polymorphism attenuates cellular antiviral responses and is associated with accelerated disease progression in idiopathic pulmonary fibrosis (IPF). The role of TLR3 L412F in bacterial infection in IPF or in acute exacerbations (AE) has not been reported. Objectives: To characterize the association between TLR3 L412F and AE-related death in IPF. To determine the effect of TLR3 L412F on the lung microbiome and on antibacterial TLR responses of primary lung fibroblasts from patients with IPF. Methods: TLR-mediated antibacterial and antiviral responses were quantitated in L412F wild-type and 412F-heterozygous primary lung fibroblasts from patients with IPF using ELISA, Western blot analysis, and quantitative PCR. Hierarchical heatmap analysis was employed to establish bacterial and viral clustering in nasopharyngeal lavage samples from patients with AE-IPF. 16S ribosomal RNA quantitative PCR and pyrosequencing were used to determine the effect of TLR3 L412F on the IPF lung microbiome. Measurements and Main Results: A significant increase in AE-related death in patients with 412F-variant IPF was reported. We established that 412F-heterozygous IPF lung fibroblasts have reduced antibacterial TLR responses to LPS (TLR4), Pam3CYSK4 (TLR1/2), flagellin (TLR5), and FSL-1 (TLR6/1) and have reduced responses to live Pseudomonas aeruginosa infection. Using 16S ribosomal RNA sequencing, we demonstrated that 412F-heterozygous patients with IPF have a dysregulated lung microbiome with increased frequencies of Streptococcus and Staphylococcus spp. Conclusions: This study reveals that TLR3 L412F dysregulates the IPF lung microbiome and reduces the responses of IPF lung fibroblasts to bacterial TLR agonists and live bacterial infection. These findings identify a candidate role for TLR3 L412F in viral- and bacterial-mediated AE death.


Asunto(s)
Fibrosis Pulmonar Idiopática , Receptor Toll-Like 3/genética , Antibacterianos , Antivirales , Progresión de la Enfermedad , Humanos , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/microbiología , ARN Ribosómico 16S
10.
J Biol Chem ; 297(6): 101417, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34793837

RESUMEN

SARM1 is a toll/interleukin-1 receptor -domain containing protein, with roles proposed in both innate immunity and neuronal degeneration. Murine SARM1 has been reported to regulate the transcription of chemokines in both neurons and macrophages; however, the extent to which SARM1 contributes to transcription regulation remains to be fully understood. Here, we identify differential gene expression in bone-marrow-derived macrophages (BMDMs) from C57BL/6 congenic 129 ES cell-derived Sarm1-/- mice compared with wild type (WT). However, we found that passenger genes, which are derived from the 129 donor strain of mice that flank the Sarm1 locus, confound interpretation of the results, since many of the identified differentially regulated genes come from this region. To re-examine the transcriptional role of SARM1 in the absence of passenger genes, here we generated three Sarm1-/- mice using CRISPR/Cas9. Treatment of neurons from these mice with vincristine, a chemotherapeutic drug causing axonal degeneration, confirmed SARM1's function in that process; however, these mice also showed that lack of SARM1 has no impact on transcription of genes previously shown to be affected such as chemokines. To gain further insight into SARM1 function, we generated an epitope-tagged SARM1 mouse. In these mice, we observed high SARM1 protein expression in the brain and brainstem and lower but detectable levels in macrophages. Overall, the generation of these SARM1 knockout and epitope-tagged mice has clarified that SARM1 is expressed in mouse macrophages yet has no general role in macrophage transcriptional regulation and has provided important new models to further explore SARM1 function.


Asunto(s)
Proteínas del Dominio Armadillo , Sistemas CRISPR-Cas , Proteínas del Citoesqueleto , Epítopos , Regulación de la Expresión Génica , Macrófagos/metabolismo , Transcripción Genética , Animales , Proteínas del Dominio Armadillo/biosíntesis , Proteínas del Dominio Armadillo/genética , Proteínas del Citoesqueleto/biosíntesis , Proteínas del Citoesqueleto/genética , Epítopos/genética , Epítopos/metabolismo , Ratones , Ratones Noqueados , Neuronas/metabolismo , Vincristina/metabolismo
11.
Nat Immunol ; 11(11): 997-1004, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20890285

RESUMEN

The detection of intracellular microbial DNA is critical to appropriate innate immune responses; however, knowledge of how such DNA is sensed is limited. Here we identify IFI16, a PYHIN protein, as an intracellular DNA sensor that mediates the induction of interferon-ß (IFN-ß). IFI16 directly associated with IFN-ß-inducing viral DNA motifs. STING, a critical mediator of IFN-ß responses to DNA, was recruited to IFI16 after DNA stimulation. Lowering the expression of IFI16 or its mouse ortholog p204 by RNA-mediated interference inhibited gene induction and activation of the transcription factors IRF3 and NF-κB induced by DNA and herpes simplex virus type 1 (HSV-1). IFI16 (p204) is the first PYHIN protein to our knowledge shown to be involved in IFN-ß induction. Thus, the PYHIN proteins IFI16 and AIM2 form a new family of innate DNA sensors we call 'AIM2-like receptors' (ALRs).


Asunto(s)
ADN Viral/inmunología , Inmunidad Innata , Espacio Intracelular/inmunología , Proteínas Nucleares/inmunología , Fosfoproteínas/inmunología , Animales , Línea Celular , Proteínas de Unión al ADN , Herpesvirus Humano 1/inmunología , Humanos , Interferón beta/inmunología , Interferón beta/metabolismo , Proteínas de la Membrana/inmunología , Ratones , Monocitos/inmunología , Transducción de Señal
12.
Immunity ; 39(3): 423-5, 2013 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-24054323

RESUMEN

If DNA accumulates in the cytosol, it activates innate immunity via recently described DNA sensors. In this issue of Immunity, Gehrke et al. (2013) show that oxidized DNA is resistant to degradation by TREX1 and thus has heightened immunostimulatory capacity.


Asunto(s)
Daño del ADN , ADN/metabolismo , Exodesoxirribonucleasas/metabolismo , Proteínas de la Membrana/metabolismo , Fosfoproteínas/metabolismo , Animales , Humanos
13.
Immunity ; 38(5): 870-80, 2013 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-23706668

RESUMEN

Although it has been appreciated for some years that cytosolic DNA is immune stimulatory, it is only in the past five years that the molecular basis of DNA sensing by the innate immune system has begun to be revealed. In particular it has been described how DNA induces type I interferon, central in antiviral responses and a mediator of autoimmunity. To date more than ten cytosolic receptors of DNA have been proposed, but STING is a key adaptor protein for most DNA-sensing pathways, and we are now beginning to understand the signaling mechanisms for STING. In this review we describe the recent progress in understanding signaling mechanisms activated by DNA and the relevance of DNA sensing to pathogen responses and autoimmunity. We highlight new insights gained into how and why the immune system responds to both pathogen and self DNA and define important questions that now need to be addressed in the field of innate immune activation by DNA.


Asunto(s)
Autoinmunidad/inmunología , ARN Helicasas DEAD-box/metabolismo , ADN/inmunología , Interferón Tipo I/inmunología , Proteínas de la Membrana/metabolismo , Animales , Comunicación Celular/inmunología , Citosol , Humanos , Inmunidad Innata , Transducción de Señal/inmunología
14.
Int J Mol Sci ; 23(20)2022 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-36293378

RESUMEN

The SARS-CoV-2 virus can utilize host cell proteases to facilitate cell entry, whereby the Spike (S) protein is cleaved at two specific sites to enable membrane fusion. Furin, transmembrane protease serine 2 (TMPRSS2), and cathepsin L (CatL) are the major proteases implicated, and are thus targets for anti-viral therapy. The human serpin (serine protease inhibitor) alpha-1 antitrypsin (A1AT) shows inhibitory activity for TMPRSS2, and has previously been found to suppress cell infection with SARS-CoV-2. Here, we have generated modified serpin inhibitors with increased specificity for these cellular proteases. Using SerpinB3 (SCCA-1), a cross-class inhibitor of CatL, as a scaffold, we have designed and produced reactive centre loop (RCL) variants to more specifically target both furin and TMPRSS2. Two further variants were generated by substituting the RCL P7-P1 with the spike protein S1/S2 cleavage site from either SARS-CoV-2 alpha or delta (P681R) sequences. Altered inhibitory specificity of purified recombinant proteins was verified in protease assays, with attenuated CatL inhibition and gain of furin or TMPRSS2 inhibition, as predicted, and modified serpins were shown to block S protein cleavage in vitro. Furthermore, the serpin variants were able to inhibit S-pseudoparticle entry into A549-ACE2-TMPRSS2 cells and suppress SARS-CoV-2 replication in Vero E6 cells expressing TMPRSS2. The construct designed to inhibit TMPRSS2 (B3-TMP) was most potent. It was more effective than A1AT for TMPRSS2 enzyme inhibition (with an eighteen-fold improvement in the second order inhibition rate constant) and for blocking SARS-CoV-2 viral replication. These findings advance the potential for serpin RCL mutagenesis to generate new inhibitors, and may lead to novel anti-viral biological molecules.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Serpinas , Humanos , SARS-CoV-2 , Furina/genética , Furina/metabolismo , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo , Serpinas/genética , Serpinas/farmacología , Catepsina L/metabolismo , Enzima Convertidora de Angiotensina 2 , Internalización del Virus , Antivirales/farmacología , Mutagénesis , Proteínas Recombinantes , Serina , Serina Endopeptidasas/genética
15.
Int J Mol Sci ; 23(3)2022 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-35163535

RESUMEN

The challenge of developing gene therapies for genetic forms of blindness is heightened by the heterogeneity of these conditions. However, mechanistic commonalities indicate key pathways that may be targeted in a gene-independent approach. Mitochondrial dysfunction and axon degeneration are common features of many neurodegenerative conditions including retinal degenerations. Here we explore the neuroprotective effect afforded by the absence of sterile alpha and Toll/interleukin-1 receptor motif-containing 1 (SARM1), a prodegenerative NADase, in a rotenone-induced mouse model of retinal ganglion cell loss and visual dysfunction. Sarm1 knockout mice retain visual function after rotenone insult, displaying preservation of photopic negative response following rotenone treatment in addition to significantly higher optokinetic response measurements than wild type mice following rotenone. Protection of spatial vision is sustained over time in both sexes and is accompanied by increased RGC survival and additionally preservation of axonal density in optic nerves of Sarm1-/- mice insulted with rotenone. Primary fibroblasts extracted from Sarm1-/- mice demonstrate an increased oxygen consumption rate relative to those from wild type mice, with significantly higher basal, maximal and spare respiratory capacity. Collectively, our data indicate that Sarm1 ablation increases mitochondrial bioenergetics and confers histological and functional protection in vivo in the mouse retina against mitochondrial dysfunction, a hallmark of many neurodegenerative conditions including a variety of ocular disorders.


Asunto(s)
Proteínas del Dominio Armadillo/genética , Proteínas del Citoesqueleto/genética , Fibroblastos/metabolismo , Degeneración Retiniana/prevención & control , Células Ganglionares de la Retina/fisiología , Rotenona/efectos adversos , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Metabolismo Energético , Femenino , Fibroblastos/citología , Técnicas de Inactivación de Genes , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Consumo de Oxígeno , Cultivo Primario de Células , Degeneración Retiniana/inducido químicamente , Degeneración Retiniana/genética
16.
J Biol Chem ; 295(14): 4438-4450, 2020 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-32102850

RESUMEN

Animal cells use pattern-recognition receptors (PRRs) to detect specific pathogens. Pathogen detection mounts an appropriate immune response, including interferon and cytokine induction. The intracellular PRR-signaling pathways that detect DNA viruses have been characterized, particularly in myeloid cells. In these pathways, cGMP-AMP synthase (cGAS) and the pyrin and HIN domain family member (PYHIN) protein interferon-γ-inducible protein 16 (IFI16) detect DNA and signal via stimulator of interferon genes protein (STING). However, although airway epithelial cells are frontline sentinels in detecting pathogens, information on how they respond to DNA viruses is limited, and the roles of PYHIN proteins in these cells are unknown. Here, we examined expression and activities of cGAS, STING, and PYHINs in human lung epithelial cells. A549 epithelial cells, commonly used for RNA-sensing studies, failed to respond to DNA because they lacked STING expression, and ectopic STING expression restored a cGAS-dependent DNA response in these cells. In contrast, NuLi-1 immortalized human bronchial epithelial cells did express STING, which was activated after DNA stimulation and mediated DNA-dependent gene induction. PYHIN1, which like IFI16 has been proposed to be a viral DNA sensor, was the only PYHIN protein expressed in both airway epithelial cell types. However, rather than having a role in DNA sensing, PYHIN1 induced proinflammatory cytokines in response to interleukin-1 (IL-1) or tumor necrosis factor α (TNFα) stimulation. Of note, PYHIN1, via its HIN domain, directly induced IL-6 and TNFα transcription, revealing that PYHIN proteins play a role in proinflammatory gene induction in airway epithelial cells.


Asunto(s)
Citocinas/metabolismo , ADN Viral/metabolismo , Inmunidad Innata , Proteínas Nucleares/metabolismo , Línea Celular , Células Epiteliales/citología , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiología , Humanos , Interleucina-1/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Nucleotidiltransferasas/antagonistas & inhibidores , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Fosfoproteínas/metabolismo , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Virus Sendai/genética , Virus Sendai/fisiología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
18.
Immunity ; 36(4): 561-71, 2012 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-22483801

RESUMEN

Recognition of DNA by the innate immune system is central to antiviral and antibacterial defenses, as well as an important contributor to autoimmune diseases involving self DNA. AIM2 (absent in melanoma 2) and IFI16 (interferon-inducible protein 16) have been identified as DNA receptors that induce inflammasome formation and interferon production, respectively. Here we present the crystal structures of their HIN domains in complex with double-stranded (ds) DNA. Non-sequence-specific DNA recognition is accomplished through electrostatic attraction between the positively charged HIN domain residues and the dsDNA sugar-phosphate backbone. An intramolecular complex of the AIM2 Pyrin and HIN domains in an autoinhibited state is liberated by DNA binding, which may facilitate the assembly of inflammasomes along the DNA staircase. These findings provide mechanistic insights into dsDNA as the activation trigger and oligomerization platform for the assembly of large innate signaling complexes such as the inflammasomes.


Asunto(s)
ADN Forma B/metabolismo , Proteínas de Unión al ADN/química , Inflamasomas/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Secuencia de Aminoácidos , Línea Celular , Cristalografía por Rayos X , ADN Forma B/química , ADN Forma B/inmunología , Humanos , Inmunidad Innata , Inflamasomas/genética , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas Nucleares/química , Unión Proteica , Pliegue de Proteína , Estructura Terciaria de Proteína , Transducción de Señal
19.
J Biol Chem ; 294(13): 5228-5229, 2019 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-30926761

RESUMEN

Poxviruses have evolved efficient proteins that bind mammalian cytokines and chemokines to suppress host immunity. Here Pontejo et al. examine in detail how one such poxviral protein, CrmD, that has activity against both mammalian tumor necrosis factor and chemokines, interacts with its host targets. They apply their findings to refine a human anti-cytokine therapeutic and increase its specificity, providing an elegant example of the benefits of mining viral proteins for therapeutically useful information.


Asunto(s)
Antiinflamatorios/inmunología , Antiinflamatorios/farmacología , Citocinas/antagonistas & inhibidores , Poxviridae/inmunología , Proteínas Virales/inmunología , Proteínas Virales/farmacología , Animales , Antiinflamatorios/química , Citocinas/inmunología , Descubrimiento de Drogas , Humanos , Poxviridae/química , Infecciones por Poxviridae/virología , Inhibidores del Factor de Necrosis Tumoral , Factores de Necrosis Tumoral/inmunología , Proteínas Virales/química
20.
J Biol Chem ; 294(45): 17031-17042, 2019 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-31558608

RESUMEN

Pathogen activation of innate immune pattern recognition receptors (PRRs) such as Toll-like receptors (TLRs) stimulates cellular signaling pathways. This often leads to outcomes that contribute to pathogen clearance. Alternatively, activation of specific PRR pathways can aid pathogen survival. The human pathogen Staphylococcus aureus is a case in point, employing strategies to escape innate immune recognition and killing by the host. As for other bacteria, PRR-stimulated type I interferon (IFN-I) induction has been proposed as one such immune escape pathway that may favor S. aureus Cell wall components of S. aureus elicit TLR2-dependent cellular responses, but the exact signaling pathways activated by S. aureus-TLR2 engagement and the consequences of their activation for the host and bacterium are not fully known. We previously showed that TLR2 activates both a cytoplasmic and an endosome-dependent signaling pathway, the latter leading to IFN-I production. Here, we demonstrate that S. aureus infection of human monocytes activates a TLR2-dependent endosomal signaling pathway, leading to IFN-I induction. We mapped the signaling components of this pathway and identified roles in IFN-I stimulation for the Toll-interleukin-1 receptor (TIR) adaptor Myd88 adaptor-like (Mal), TNF receptor-associated factor 6 (TRAF6), and IκB kinase (IKK)-related kinases, but not for TRIF-related adaptor molecule (TRAM) and TRAF3. Importantly, monocyte TLR2-dependent endosomal signaling enabled immune escape for S. aureus, because this pathway, but not IFN-I per se, contributed to intracellular bacterial survival. These results reveal a TLR2-dependent mechanism in human monocytes whereby S. aureus manipulates innate immune signaling for its survival in cells.


Asunto(s)
Endosomas/metabolismo , Interferón Tipo I/metabolismo , Viabilidad Microbiana , Monocitos/microbiología , Transducción de Señal , Staphylococcus aureus/fisiología , Receptor Toll-Like 2/metabolismo , Animales , Línea Celular , Humanos , Quinasa I-kappa B/metabolismo , Ratones , Monocitos/citología , Factor 6 Asociado a Receptor de TNF/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA