Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 54(9): 1909-1911, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34525334

RESUMEN

Some RNAs can assume a Z conformation, an unusual, left-handed turn. In this issue of Immunity, three studies report that mutations in the Zα-RNA binding domain of the adenosine deaminase ADAR1 are sufficient to induce autoinflammatory disease in mice, which models human Aicardí-Goutières syndrome, highlighting the important role of Z-RNA editing in limiting innate immune recognition of endogenous RNA.


Asunto(s)
Enfermedades Autoinmunes del Sistema Nervioso , ARN , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Animales , Ratones , ARN/genética , Edición de ARN , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
2.
Immunity ; 53(1): 54-77, 2020 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-32668228

RESUMEN

All lifeforms have developed highly sophisticated systems equipped to detect altered self and non-self nucleic acids (NA). In vertebrates, NA-sensing receptors safeguard the integrity of the organism by detecting pathogens, dyshomeostasis and damage, and inducing appropriate responses to eliminate pathogens and reconstitute homeostasis. Effector mechanisms include i) immune signaling, ii) restriction of NA functions such as inhibition of mRNA translation, and iii) cell death pathways. An appropriate effector response is necessary for host defense, but dysregulated NA-sensing can lead to devastating autoimmune and autoinflammatory disease. Their inherent biochemical similarity renders the reliable distinction between self NA under homeostatic conditions and altered or exogenous NA particularly challenging. In this review, we provide an overview of recent progress in our understanding of the closely coordinated and regulated network of innate immune receptors, restriction factors, and nucleases to effectively respond to pathogens and maintain host integrity.


Asunto(s)
Autoantígenos/inmunología , Autoinmunidad/inmunología , Ácidos Nucleicos/inmunología , Receptores Inmunológicos/inmunología , Animales , Enfermedades Autoinmunes/inmunología , Humanos , Inmunidad Innata/inmunología , Inflamasomas/inmunología , Inflamasomas/metabolismo , Transducción de Señal/inmunología
3.
Immunity ; 52(4): 591-605.e6, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32294405

RESUMEN

Human toll-like receptor 8 (TLR8) activation induces a potent T helper-1 (Th1) cell response critical for defense against intracellular pathogens, including protozoa. The receptor harbors two distinct binding sites, uridine and di- and/or trinucleotides, but the RNases upstream of TLR8 remain poorly characterized. We identified two endolysosomal endoribonucleases, RNase T2 and RNase 2, that act synergistically to release uridine from oligoribonucleotides. RNase T2 cleaves preferentially before, and RNase 2 after, uridines. Live bacteria, P. falciparum-infected red blood cells, purified pathogen RNA, and synthetic oligoribonucleotides all required RNase 2 and T2 processing to activate TLR8. Uridine supplementation restored RNA recognition in RNASE2-/- or RNASET2-/- but not RNASE2-/-RNASET2-/- cells. Primary immune cells from RNase T2-hypomorphic patients lacked a response to bacterial RNA but responded robustly to small-molecule TLR8 ligands. Our data identify an essential function of RNase T2 and RNase 2 upstream of TLR8 and provide insight into TLR8 activation.


Asunto(s)
Endorribonucleasas/metabolismo , Monocitos/inmunología , Neutrófilos/inmunología , ARN Bacteriano/metabolismo , ARN Protozoario/metabolismo , Receptor Toll-Like 8/metabolismo , Sistemas CRISPR-Cas , Línea Celular , Endorribonucleasas/inmunología , Eritrocitos/inmunología , Eritrocitos/parasitología , Escherichia coli/química , Escherichia coli/inmunología , Edición Génica/métodos , Humanos , Listeria monocytogenes/química , Listeria monocytogenes/inmunología , Monocitos/microbiología , Monocitos/parasitología , Neutrófilos/microbiología , Neutrófilos/parasitología , Plasmodium falciparum/química , Plasmodium falciparum/inmunología , Cultivo Primario de Células , Estabilidad del ARN , ARN Bacteriano/inmunología , ARN Protozoario/inmunología , Serratia marcescens/química , Serratia marcescens/inmunología , Staphylococcus aureus/química , Staphylococcus aureus/inmunología , Streptococcus/química , Streptococcus/inmunología , Células THP-1 , Receptor Toll-Like 8/inmunología
4.
Nat Immunol ; 16(10): 1025-33, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26343537

RESUMEN

Cytosolic DNA that emerges during infection with a retrovirus or DNA virus triggers antiviral type I interferon responses. So far, only double-stranded DNA (dsDNA) over 40 base pairs (bp) in length has been considered immunostimulatory. Here we found that unpaired DNA nucleotides flanking short base-paired DNA stretches, as in stem-loop structures of single-stranded DNA (ssDNA) derived from human immunodeficiency virus type 1 (HIV-1), activated the type I interferon-inducing DNA sensor cGAS in a sequence-dependent manner. DNA structures containing unpaired guanosines flanking short (12- to 20-bp) dsDNA (Y-form DNA) were highly stimulatory and specifically enhanced the enzymatic activity of cGAS. Furthermore, we found that primary HIV-1 reverse transcripts represented the predominant viral cytosolic DNA species during early infection of macrophages and that these ssDNAs were highly immunostimulatory. Collectively, our study identifies unpaired guanosines in Y-form DNA as a highly active, minimal cGAS recognition motif that enables detection of HIV-1 ssDNA.


Asunto(s)
ADN Complementario/química , ADN Viral/química , ADN Viral/inmunología , VIH-1/genética , VIH-1/inmunología , Interferón-alfa/inmunología , Nucleotidiltransferasas/genética , Animales , Línea Celular , Células Cultivadas , ADN Complementario/genética , ADN Complementario/inmunología , ADN Viral/genética , Células HEK293 , Humanos , Inmunización , Ratones
5.
Nucleic Acids Res ; 51(21): 11893-11910, 2023 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-37831086

RESUMEN

RIG-I is a cytosolic receptor of viral RNA essential for the immune response to numerous RNA viruses. Accordingly, RIG-I must sensitively detect viral RNA yet tolerate abundant self-RNA species. The basic binding cleft and an aromatic amino acid of the RIG-I C-terminal domain(CTD) mediate high-affinity recognition of 5'triphosphorylated and 5'base-paired RNA(dsRNA). Here, we found that, while 5'unmodified hydroxyl(OH)-dsRNA demonstrated residual activation potential, 5'-monophosphate(5'p)-termini, present on most cellular RNAs, prevented RIG-I activation. Determination of CTD/dsRNA co-crystal structures and mutant activation studies revealed that the evolutionarily conserved I875 within the CTD sterically inhibits 5'p-dsRNA binding. RIG-I(I875A) was activated by both synthetic 5'p-dsRNA and endogenous long dsRNA within the polyA-rich fraction of total cellular RNA. RIG-I(I875A) specifically interacted with long, polyA-bearing, mitochondrial(mt) RNA, and depletion of mtRNA from total RNA abolished its activation. Altogether, our study demonstrates that avoidance of 5'p-RNA recognition is crucial to prevent mtRNA-triggered RIG-I-mediated autoinflammation.


Asunto(s)
Proteína 58 DEAD Box , Isoleucina , Receptores Inmunológicos , Proteína 58 DEAD Box/química , Proteína 58 DEAD Box/genética , Proteína 58 DEAD Box/metabolismo , Tolerancia Inmunológica , Isoleucina/genética , ARN Bicatenario/genética , ARN Mitocondrial/genética , ARN Mitocondrial/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Humanos , Receptores Inmunológicos/química , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo
6.
Immunity ; 43(1): 41-51, 2015 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-26187414

RESUMEN

The cytosolic helicase retinoic acid-inducible gene-I (RIG-I) initiates immune responses to most RNA viruses by detecting viral 5'-triphosphorylated RNA (pppRNA). Although endogenous mRNA is also 5'-triphosphorylated, backbone modifications and the 5'-ppp-linked methylguanosine ((m7)G) cap prevent immunorecognition. Here we show that the methylation status of endogenous capped mRNA at the 5'-terminal nucleotide (N1) was crucial to prevent RIG-I activation. Moreover, we identified a single conserved amino acid (H830) in the RIG-I RNA binding pocket as the mediator of steric exclusion of N1-2'O-methylated RNA. H830A alteration (RIG-I(H830A)) restored binding of N1-2'O-methylated pppRNA. Consequently, endogenous mRNA activated the RIG-I(H830A) mutant but not wild-type RIG-I. Similarly, knockdown of the endogenous N1-2'O-methyltransferase led to considerable RIG-I stimulation in the absence of exogenous stimuli. Studies involving yellow-fever-virus-encoded 2'O-methyltransferase and RIG-I(H830A) revealed that viruses exploit this mechanism to escape RIG-I. Our data reveal a new role for cap N1-2'O-methylation in RIG-I tolerance of self-RNA.


Asunto(s)
ARN Helicasas DEAD-box/genética , Tolerancia Inmunológica/genética , Procesamiento Postranscripcional del ARN/genética , ARN/genética , Virus de la Fiebre Amarilla/enzimología , Secuencia de Aminoácidos , Animales , Células Cultivadas , Proteína 58 DEAD Box , Activación Enzimática/genética , Activación Enzimática/inmunología , Histidina/genética , Humanos , Metilación , Metiltransferasas/genética , Ratones , Estructura Terciaria de Proteína , ARN/química , ARN/inmunología , ARN Viral/inmunología , Receptores Inmunológicos , Virus de la Fiebre Amarilla/genética
7.
Int J Mol Sci ; 24(15)2023 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-37569596

RESUMEN

Immune surveillance by natural killer (NK) cells and their recruitment to sites of inflammation renders them susceptible to viral infection, potentially modulating their effector function. Here, we analyzed innate RNA receptor signaling in NK cells downstream of direct Influenza A virus (IAV) infection and its impact on NK cell effector function. Infection of NK cells with IAV resulted in the activation of TBK1, NF-Ï°B and subsequent type-I IFN secretion. CRISPR-generated knockouts in primary human NK cells revealed that this effect depended on the antiviral cytosolic RNA receptor RIG-I. Transfection of NK cells with synthetic 3p-dsRNA, a strong RIG-I agonist that mimics viral RNA, resulted in a similar phenotype and rendered NK cells resistant to subsequent IAV infection. Strikingly, both IAV infection and 3p-dsRNA transfection enhanced degranulation and cytokine production by NK cells when exposed to target cells. Thus, RIG-I activation in NK cells both supports their cell intrinsic viral defense and enhances their cytotoxic effector function against target cells.


Asunto(s)
Virus de la Influenza A , Gripe Humana , Interferón Tipo I , Humanos , Virus de la Influenza A/fisiología , Células Asesinas Naturales , ARN
8.
J Clin Immunol ; 42(2): 325-335, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34783940

RESUMEN

PURPOSE: NLRC4-associated autoinflammatory disease (NLRC4-AID) is an autosomal dominant condition presenting with a range of clinical manifestations which can include macrophage activation syndrome (MAS) and severe enterocolitis. We now report the first homozygous mutation in NLRC4 (c.478G > A, p.A160T) causing autoinflammatory disease with immune dysregulation and find that heterozygous carriers in the general population are at increased risk of developing ulcerative colitis. METHODS: Circulating immune cells and inflammatory markers were profiled and historical clinical data interrogated. DNA was extracted and sequenced using standard procedures. Inflammasome activation assays for ASC speck formation, pyroptosis, and IL-1ß/IL-18 secretion confirmed pathogenicity of the mutation in vitro. Genome-wide association of NLRC4 (A160T) with ulcerative colitis was examined using data from the IBD exomes portal. RESULTS: A 60-year-old Brazilian female patient was evaluated for recurrent episodes of systemic inflammation from six months of age. Episodes were characterized by recurrent low-grade fever, chills, oral ulceration, uveitis, arthralgia, and abdominal pain, followed by diarrhea with mucus and variable skin rash. High doses of corticosteroids were somewhat effective in controlling disease and anti-IL-1ß therapy partially controlled symptoms. While on treatment, serum IL-1ß and IL-18 levels remained elevated. Genetic investigations identified a homozygous mutation in NLRC4 (A160T), inherited in a recessive fashion. Increased ASC speck formation and IL-1ß/IL-18 secretion confirmed pathogenicity when NLRC4 (A160T) was analyzed in human cell lines. This allele is significantly enriched in patients with ulcerative colitis: OR 2.546 (95% 1.778-3.644), P = 0.01305. CONCLUSION: NLRC4 (A160T) can either cause recessively inherited autoinflammation and immune dysregulation, or function as a heterozygous risk factor for the development of ulcerative colitis.


Asunto(s)
Colitis Ulcerosa , Enfermedades Autoinflamatorias Hereditarias , Proteínas Adaptadoras de Señalización CARD/genética , Proteínas Adaptadoras de Señalización CARD/metabolismo , Proteínas de Unión al Calcio/genética , Colitis Ulcerosa/diagnóstico , Colitis Ulcerosa/genética , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Inflamasomas/metabolismo , Persona de Mediana Edad
9.
J Med Virol ; 94(1): 388-392, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34415572

RESUMEN

In the current COVID-19 pandemic, a better understanding of the relationship between merely binding and functionally neutralizing antibodies is necessary to characterize protective antiviral immunity following infection or vaccination. This study analyzes the level of correlation between the novel quantitative EUROIMMUN Anti-SARS-CoV-2 QuantiVac ELISA (IgG) and a microneutralization assay. A panel of 123 plasma samples from a COVID-19 outbreak study population, preselected by semiquantitative anti-SARS-CoV-2 IgG testing, was used to assess the relationship between the novel quantitative ELISA (IgG) and a microneutralization assay. Binding IgG targeting the S1 antigen was detected in 106 (86.2%) samples using the QuantiVac ELISA, while 89 (72.4%) samples showed neutralizing antibody activity. Spearman's correlation analysis demonstrated a strong positive relationship between anti-S1 IgG levels and neutralizing antibody titers (rs = 0.819, p < 0.0001). High and low anti-S1 IgG levels were associated with a positive predictive value of 72.0% for high-titer neutralizing antibodies and a negative predictive value of 90.8% for low-titer neutralizing antibodies, respectively. These results substantiate the implementation of the QuantiVac ELISA to assess protective immunity following infection or vaccination.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Ensayo de Inmunoadsorción Enzimática/métodos , Inmunoglobulina G/sangre , SARS-CoV-2/inmunología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , COVID-19/inmunología , COVID-19/patología , Prueba Serológica para COVID-19/métodos , Niño , Preescolar , Femenino , Humanos , Masculino , Persona de Mediana Edad , Pruebas de Neutralización/métodos , Sensibilidad y Especificidad , Glicoproteína de la Espiga del Coronavirus/inmunología , Adulto Joven
10.
Int J Mol Sci ; 23(19)2022 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-36232437

RESUMEN

Recognition of RNA by receptors of the innate immune system is regulated by various posttranslational modifications. Different single 2'-O-ribose (2'-O-) methylations have been shown to convert TLR7/TLR8 ligands into specific TLR8 ligands, so we investigated whether the position of 2'-O-methylation is crucial for its function. To this end, we designed different 2'-O-methylated RNA oligoribonucleotides (ORN), investigating their immune activity in various cell systems and analyzing degradation under RNase T2 treatment. We found that the 18S rRNA-derived TLR7/8 ligand, RNA63, was differentially digested as a result of 2'-O-methylation, leading to variations in TLR8 and TLR7 inhibition. The suitability of certain 2'-O-methylated RNA63 derivatives as TLR8 agonists was further demonstrated by the fact that other RNA sequences were only weak TLR8 agonists. We were thus able to identify specific 2'-O-methylated RNA derivatives as optimal TLR8 ligands.


Asunto(s)
Receptor Toll-Like 7 , Receptor Toll-Like 8 , Ligandos , Metilación , Oligorribonucleótidos/metabolismo , Procesamiento Proteico-Postraduccional , ARN/metabolismo , ARN Ribosómico 18S/metabolismo , Ribosa , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 8/metabolismo
11.
Int J Cancer ; 144(7): 1645-1656, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30230526

RESUMEN

Activation of the innate immune receptor retinoic acid-inducible gene I (RIG-I) by its specific ligand 5'-triphosphate RNA (3pRNA) triggers anti-tumor immunity, which is dependent on natural killer (NK) cell activation and cytokine induction. However, to date, RIG-I expression and the functional consequences of RIG-I activation in NK cells have not been examined. Here, we show for the first time the expression of RIG-I in human NK cells and their activation upon RIG-I ligand (3pRNA) transfection. 3pRNA-activated NK cells killed melanoma cells more efficiently than NK cells activated by type I interferon. Stimulation of RIG-I in NK cells specifically increased the surface expression of membrane-bound TNF-related apoptosis-inducing ligand (TRAIL) on NK cells, while activated NK cell receptors were not affected. RIG-I-induced membrane-bound TRAIL initiated death-receptor-pathway-mediated apoptosis not only in allogeneic but also in autologous human leukocyte antigen (HLA) class I-positive and HLA class I-negative melanoma cells. These results identify the direct activation of RIG-I in NK cells as a novel mechanism for how RIG-I can trigger enhanced NK cell killing of tumor cells, underscoring the potential of RIG-I activation for tumor immunotherapy.


Asunto(s)
Proteína 58 DEAD Box/metabolismo , Células Asesinas Naturales/citología , Melanoma/inmunología , Melanoma/terapia , ARN/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Apoptosis , Técnicas de Cocultivo , Citotoxicidad Inmunológica , Humanos , Células Asesinas Naturales/inmunología , Ligandos , Proteína 1 de la Membrana Asociada a los Lisosomas/metabolismo , ARN/genética , Receptores Inmunológicos , Transfección , Trasplante Autólogo , Células Tumorales Cultivadas
12.
Med Microbiol Immunol ; 206(2): 93-103, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27832373

RESUMEN

Human papillomaviruses (HPVs) are an acknowledged cause of a subset of oropharyngeal cancers, especially of tonsillar cancer. Similar to HPV, some human polyomaviruses (HPyVs), such as Merkel cell polyomavirus (MCPyV), have an oncogenic potential. Recently, several novel HPyVs have been discovered. The aim of our study was to determine viral DNA prevalence and viral DNA load of 13 different HPyVs in benign and malignant tonsillar tissue and to compare the data with those found for HPV. A total of 78 biopsies of palatine tonsils with a histologic diagnosis of non-malignant disease (chronic tonsillitis, tonsillar hyperplasia, n = 40) or tonsillar squamous cell carcinoma (n = 38) were included in the study. HPyV DNA prevalence and viral load were determined by virus-specific quantitative real-time PCRs. JCPyV (1/40, 2.5%) and WUPyV (3/40, 7.5%) were only found in non-malignant tonsillar tissue. HPyV7 and HPyV10 were only detected in one (2.6%) and seven (18.4%) of the 38 cancer biopsies, respectively. Both MCPyV (8/38, 21.1 vs. 4/40, 10.0%) and HPyV6 (2/38, 5.3 vs. 1/40, 2.5%) were found more frequently in cancer samples than in non-malignant tissue, but the differences were not significant. BKPyV, KIPyV, TSPyV, HPyV9, STLPyV, HPyV12 and NJPyV were not discovered in any of the samples. HPyV loads found in HPyV DNA-positive biopsies were very low with no difference between non-malignant and malignant samples (median load <0.0001 HPyV DNA copies per beta-globin gene copy, respectively). In contrast to HPyV, high-risk HPV types (HPV16/HPV18) were found significantly more frequently in tonsillar cancers than in non-malignant tonsillar tissue (17/38, 44.7 vs. 2/40, 5.0%, p < 0.001). Furthermore, high-risk HPV DNA loads were significantly higher in the cancer compared to the non-malignant samples (median load 11.861 vs. 7 × 10-6 HPV DNA copies per beta-globin gene copy, p = 0.012). While both HPV and HPyV may persist in tonsillar tissue, our data on HPyV DNA prevalence and load do not support a role of HPyV in tonsillar carcinogenesis, neither alone nor as co-infecting agents of HPV.


Asunto(s)
Tonsila Palatina/patología , Tonsila Palatina/virología , Papillomaviridae , Infecciones por Papillomavirus/complicaciones , Infecciones por Papillomavirus/virología , Infecciones por Polyomavirus/complicaciones , Infecciones por Polyomavirus/virología , Poliomavirus , Neoplasias Tonsilares/epidemiología , Neoplasias Tonsilares/etiología , Adulto , Anciano , Biopsia , Carcinoma , ADN Viral , Femenino , Humanos , Hiperplasia , Masculino , Persona de Mediana Edad , Papillomaviridae/genética , Poliomavirus/genética , Prevalencia , Estudios Retrospectivos , Factores de Riesgo , Neoplasias Tonsilares/diagnóstico , Infecciones Tumorales por Virus/complicaciones , Infecciones Tumorales por Virus/virología , Carga Viral , Adulto Joven
13.
Kidney Int ; 90(3): 525-39, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27262364

RESUMEN

Intrarenal crystal formation activates the Nlrp3 inflammasome in myeloid cells and triggers a profound inflammatory response. Here, we studied whether a specific inhibitor of the Nlrp3 inflammasome, CP-456,773, can prevent kidney fibrosis in a murine model of crystal nephropathy induced by diets rich in oxalate or adenine. Inflammasome activation in renal dendritic cells and the resulting interleukin (IL)-1ß and IL-18 production were markedly reduced by CP-456,773 treatment both ex vivo and in vivo. We directly visualized intrarenal inflammasome activation and its inhibition by CP-456,773 in vivo by adoptive transfer of bone marrow cells transduced with interleukin-1ß-Gaussia luciferase, a proteolytic luciferase-based reporter for inflammasome activation, into irradiated mice. CP-456,773 treatment strongly attenuated kidney fibrosis when given early in the genesis of crystal nephropathy, but was unable to reverse established crystal-induced fibrosis. The urinary IL-18 concentration appeared to be a useful noninvasive biomarker for renal inflammasome activation. Finally, NLRP3 inhibition did not compromise adaptive immune responses as previously reported for the global inhibition of IL-1 signaling. Thus, early NLRP3 inhibition by CP-456,773 may be an effective treatment for crystal nephropathy. Use of iGLuc transfected cells introduces a novel imaging technique for inflammasome activation in mice.


Asunto(s)
Células Dendríticas/metabolismo , Compuestos Heterocíclicos de 4 o más Anillos/uso terapéutico , Inflamasomas/efectos de los fármacos , Riñón/patología , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Nefritis/tratamiento farmacológico , Sulfonas/uso terapéutico , Adenina/efectos adversos , Traslado Adoptivo , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Fibrosis , Furanos , Humanos , Inmunohistoquímica , Indenos , Inflamasomas/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Riñón/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Nefritis/inducido químicamente , Oxalatos/efectos adversos , Cultivo Primario de Células , Transducción de Señal , Sulfonamidas
14.
Eur J Immunol ; 45(10): 2911-7, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26174085

RESUMEN

Inflammasome activation culminates in activation of caspase-1, which leads to the maturation and subsequent release of cytokines of the interleukin 1 (IL-1) family and results in a particular form of cell death known as pyroptosis. In addition, in the murine system, a so-called non-canonical inflammasome involving caspase-11 has been described that directly responds to cytosolic LPS. Here, we show that the human monocytic cell line THP1 activates the inflammasome in response to cytosolic LPS in a TLR4-independent fashion. This response is mediated by caspase-4 and accompanied by caspase-1 activation, pyroptosis, and IL-1ß maturation. In addition to caspase-4, efficient IL-1ß conversion upon intracellular LPS delivery relies on potassium efflux, NLRP3, ASC, and caspase-1, indicating that although caspase-4 activation alone is sufficient to induce pyroptosis, this process depends on the NLRP3 inflammasome activation to drive IL-1ß maturation. Altogether, this study provides evidence for the presence of a non-canonical inflammasome in humans and its dependence on caspase-4.


Asunto(s)
Proteínas Portadoras/inmunología , Caspasas Iniciadoras/inmunología , Inflamasomas/inmunología , Células Mieloides/inmunología , Proteínas Portadoras/genética , Caspasa 1/genética , Caspasa 1/inmunología , Caspasas Iniciadoras/genética , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Humanos , Inflamasomas/genética , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Lipopolisacáridos/farmacología , Células Mieloides/citología , Proteína con Dominio Pirina 3 de la Familia NLR
15.
Nat Methods ; 10(2): 147-154, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23291722

RESUMEN

Measurement of protease activity in living cells or organisms remains a challenging task. We here present a transgene-encoded biosensor that reports the proteolytic activity of caspase-1 in the course of inflammasome activation and that of other proteases in a highly sensitive and specific manner. This protease reporter is based on the biological activity of a pro-interleukin (IL)-1ß-Gaussia luciferase (iGLuc) fusion construct, in which pro-IL-1ß-dependent formation of protein aggregates renders GLuc enzyme inactive. Cleavage leads to monomerization of this biosensor protein, resulting in a strong gain in luciferase activity. Exchange of the canonical caspase-1 cleavage site in this reporter construct allows the generation of protease biosensors with additional specificities. The high sensitivity, signal-to-background ratio and specificity of the iGLuc system renders it a useful tool to study proteolytic events in mouse and human cells at high throughput and to monitor protease activity in mice in vivo.


Asunto(s)
Técnicas Biosensibles/métodos , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Luciferasas/metabolismo , Animales , Caspasa 1/metabolismo , Línea Celular , Activación Enzimática , Humanos , Ratones , Transfección
16.
Nat Commun ; 15(1): 1534, 2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-38378748

RESUMEN

Myotonic dystrophy type 2 (DM2) is a tetranucleotide CCTG repeat expansion disease associated with an increased prevalence of autoimmunity. Here, we identified an elevated type I interferon (IFN) signature in peripheral blood mononuclear cells and primary fibroblasts of DM2 patients as a trigger of chronic immune stimulation. Although RNA-repeat accumulation was prevalent in the cytosol of DM2-patient fibroblasts, type-I IFN release did not depend on innate RNA immune sensors but rather the DNA sensor cGAS and the prevalence of mitochondrial DNA (mtDNA) in the cytoplasm. Sublethal mtDNA release was promoted by a chronic activation of the ATF6 branch of the unfolded protein response (UPR) in reaction to RNA-repeat accumulation and non-AUG translated tetrapeptide expansion proteins. ATF6-dependent mtDNA release and resulting cGAS/STING activation could also be recapitulated in human THP-1 monocytes exposed to chronic endoplasmic reticulum (ER) stress. Altogether, our study demonstrates a novel mechanism by which large repeat expansions cause chronic endoplasmic reticulum stress and associated mtDNA leakage. This mtDNA is, in turn, sensed by the cGAS/STING pathway and induces a type-I IFN response predisposing to autoimmunity. Elucidating this pathway reveals new potential therapeutic targets for autoimmune disorders associated with repeat expansion diseases.


Asunto(s)
Enfermedades Autoinmunes , Interferón Tipo I , Distrofia Miotónica , Humanos , Distrofia Miotónica/genética , Distrofia Miotónica/metabolismo , ADN Mitocondrial/genética , Autoinmunidad/genética , Leucocitos Mononucleares/metabolismo , ARN , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Estrés del Retículo Endoplásmico/genética
17.
J Immunol ; 187(2): 613-7, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21677136

RESUMEN

A common denominator among the multiple damage-inducing agents that ultimately lead to activation of NLRP3 has not yet been identified. Recently, production of reactive oxygen species (ROS) has been suggested to act as a common event upstream of the NLRP3 inflammasome machinery. Because de novo translation of NLRP3 is an essential step in the activation of NLRP3, we investigated the role of substances that inhibit either ROS production or its oxidative activity. Although we observe that NLRP3 inflammasome activation is unique among other known inflammasomes in its sensitivity to ROS inhibition, we have found that this phenomenon is attributable to the fact that NLRP3 strictly requires priming by a proinflammatory signal, a step that is blocked by ROS inhibitors. Although these data do not exclude a general role for ROS production in the process of NLRP3-triggered inflammation, they would put ROS upstream of NLRP3 induction, but not activation.


Asunto(s)
Proteínas Portadoras/antagonistas & inhibidores , Regulación hacia Abajo/inmunología , Inflamasomas/antagonistas & inhibidores , Inflamasomas/metabolismo , Activación de Linfocitos/inmunología , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Animales , Compuestos de Bifenilo/farmacología , Proteínas Portadoras/fisiología , Células Cultivadas , Inflamasomas/deficiencia , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Glicoproteínas de Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , NADPH Oxidasa 2 , NADPH Oxidasas , Proteína con Dominio Pirina 3 de la Familia NLR , Compuestos Onio/farmacología , Especies Reactivas de Oxígeno/metabolismo , Especies Reactivas de Oxígeno/farmacología , Factor de Transcripción STAT1 , Transducción de Señal/inmunología
18.
Cells ; 12(2)2023 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-36672163

RESUMEN

Epilepsy and mental retardation are known to be associated with pathogenic mutations in a broad range of genes that are expressed in the brain and have a role in neurodevelopment. Here, we report on a family with three affected individuals whose clinical symptoms closely resemble a neurodevelopmental disorder. Whole-exome sequencing identified a homozygous stop-gain mutation, p.Gln19*, in the BATF2 gene in the patients. The BATF2 transcription factor is predominantly expressed in macrophages and monocytes and has been reported to modulate AP-1 transcription factor-mediated pro-inflammatory responses. Transcriptome analysis showed altered base-level expression of interferon-stimulated genes in the patients' blood, typical for type I interferonopathies. Peripheral blood mononuclear cells from all three patients demonstrated elevated responses to innate immune stimuli, which could be reproduced in CRISPR-Cas9-generated BATF2-/- human monocytic cell lines. BATF2 is, therefore, a novel disease-associated gene candidate for severe epilepsy and mental retardation related to dysregulation of immune responses, which underscores the relevance of neuroinflammation for epilepsy.


Asunto(s)
Discapacidad Intelectual , Factores de Transcripción , Humanos , Factores de Transcripción/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Leucocitos Mononucleares/metabolismo , Inmunidad , Fenotipo
19.
J Mol Cell Biol ; 15(1)2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-36626927

RESUMEN

Radiotherapy induces DNA damage, resulting in cell-cycle arrest and activation of cell-intrinsic death pathways. However, the radioresistance of some tumour entities such as malignant melanoma limits its clinical application. The innate immune sensing receptor retinoic acid-inducible gene I (RIG-I) is ubiquitously expressed and upon activation triggers an immunogenic form of cell death in a variety of tumour cell types including melanoma. To date, the potential of RIG-I ligands to overcome radioresistance of tumour cells has not been investigated. Here, we demonstrate that RIG-I activation enhanced the extent and immunogenicity of irradiation-induced tumour cell death in human and murine melanoma cells in vitro and improved survival in the murine B16 melanoma model in vivo. Transcriptome analysis pointed to a central role for p53, which was confirmed using p53-/- B16 cells. In vivo, the additional effect of RIG-I in combination with irradiation on tumour growth was absent in mice carrying p53-/- B16 tumours, while the antitumoural response to RIG-I stimulation alone was maintained. Our results identify p53 as a pivotal checkpoint that is triggered by RIG-I resulting in enhanced irradiation-induced tumour cell death. Thus, the combined administration of RIG-I ligands and radiotherapy is a promising approach to treating radioresistant tumours with a functional p53 pathway, such as melanoma.


Asunto(s)
Melanoma Experimental , Proteína p53 Supresora de Tumor , Animales , Ratones , Humanos , Proteína p53 Supresora de Tumor/genética , Línea Celular Tumoral , Ligandos , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Inmunoterapia/métodos , Melanoma Cutáneo Maligno
20.
Cell Mol Life Sci ; 68(5): 765-83, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21072676

RESUMEN

The innate immune system relies on its capability to detect invading microbes, tissue damage, or stress via evolutionarily conserved receptors. The nucleotide-binding domain leucine-rich repeat (NLR)-containing family of pattern recognition receptors includes several proteins that drive inflammation in response to a wide variety of molecular patterns. In particular, the NLRs that participate in the formation of a molecular scaffold termed the "inflammasome" have been intensively studied in past years. Inflammasome activation by multiple types of tissue damage or by pathogen-associated signatures results in the autocatalytic cleavage of caspase-1 and ultimately leads to the processing and thus secretion of pro-inflammatory cytokines, most importantly interleukin (IL)-1ß and IL-18. Here, we review the current knowledge of mechanisms leading to the activation of inflammasomes. In particular, we focus on the controversial molecular mechanisms that regulate NLRP3 signaling and highlight recent advancements in DNA sensing by the inflammasome receptor AIM2.


Asunto(s)
Inmunidad Innata , Inflamasomas/fisiología , Modelos Inmunológicos , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/fisiología , Proteínas Reguladoras de la Apoptosis/química , Proteínas Reguladoras de la Apoptosis/fisiología , Proteínas Adaptadoras de Señalización CARD/química , Proteínas Adaptadoras de Señalización CARD/fisiología , Proteínas de Unión al Calcio/química , Proteínas de Unión al Calcio/fisiología , Proteínas Portadoras/química , Proteínas Portadoras/fisiología , Caspasa 1/metabolismo , Proteínas de Unión al ADN , Humanos , Interleucina-1beta/metabolismo , Interleucina-1beta/fisiología , Proteína con Dominio Pirina 3 de la Familia NLR , Proteínas NLR , Proteínas Nucleares/química , Proteínas Nucleares/fisiología , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA