Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Clin Immunol ; 216: 108466, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32470544

RESUMEN

STING-associated vasculopathy with onset in infancy (SAVI) is an autoimmune disease caused by heterozygous gain of function mutations of STING (stimulator of interferon genes) that had initially been classified as a type I interferonopathy. We recently reported a genetically engineered mouse strain carrying a common SAVI-associated STING mutation. These STING N153S/WT mice reproduce key features of SAVI, including lung inflammation, loss of T cells in spleen and blood, splenomegaly and thymic hypoplasia. Here we show that αß T lymphocytopenia is due to disrupted T cell development and is associated with impaired T cell activation and a relative increase in γδ T cell numbers. These alterations were not rescued by additional knockout of the type I IFN receptor (IFNAR1). Collectively, our findings consolidate the concept that constitutive STING signalling leads to a SCID-like phenotype in STING N153S/WT mice.


Asunto(s)
Interferón Tipo I/inmunología , Proteínas de la Membrana/inmunología , Enfermedades Vasculares/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Mutación con Ganancia de Función/inmunología , Humanos , Inflamación/inmunología , Linfocitos Intraepiteliales/inmunología , Linfopenia/inmunología , Masculino , Ratones , Ratones SCID , Receptor de Interferón alfa y beta/inmunología
2.
J Allergy Clin Immunol ; 144(1): 254-266.e8, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30772497

RESUMEN

BACKGROUND: Monogenic interferonopathies are thought to be mediated by type I interferon. For example, a gain-of-function mutation in stimulator of interferon genes (STING; N153S) upregulates type I interferon-stimulated genes and causes perivascular inflammatory lung disease in mice. The equivalent mutation in human subjects also causes lung disease, which is thought to require signaling through the cyclic GMP-AMP synthase (cGAS)-STING pathway and subsequent activation of interferon regulatory factors (IRFs) 3 and 7, type I interferon, and interferon-stimulated genes. OBJECTIVE: We set out to define the roles of cGAS, IRF3, IRF7, the type I interferon receptor (IFN-α and IFN-ß receptor subunit 1 [IFNAR1]), T cells, and B cells in spontaneous lung disease in STING N153S mice. METHODS: STING N153S mice were crossed to animals lacking cGAS, IRF3/IRF7, IFNAR1, adaptive immunity, αß T cells, and mature B cells. Mice were evaluated for spontaneous lung disease. Additionally, bone marrow chimeric mice were assessed for lung disease severity and survival. RESULTS: Lung disease in STING N153S mice developed independently of cGAS, IRF3/IRF7, and IFNAR1. Bone marrow transplantation revealed that certain features of STING N153S-associated disease are intrinsic to the hematopoietic compartment. Recombination-activating gene 1 (Rag1)-/- STING N153S mice that lack adaptive immunity had no lung disease, and T-cell receptor ß chain (Tcrb)-/- STING N153S animals only had mild disease. STING N153S led to a reduction in percentages and numbers of naive and regulatory T cells, as well as an increased frequency of cytokine-producing effector T cells. CONCLUSION: Spontaneous lung disease in STING N153S mice develops independently of type I interferon signaling and cGAS. STING N153S relies primarily on T cells to promote lung disease in mice.


Asunto(s)
Enfermedades Pulmonares/inmunología , Proteínas de la Membrana/inmunología , Linfocitos T/inmunología , Animales , Linfocitos B/inmunología , Trasplante de Médula Ósea , Femenino , Mutación con Ganancia de Función , Interferón Tipo I/inmunología , Pulmón/inmunología , Masculino , Proteínas de la Membrana/genética , Ratones Transgénicos , Nucleotidiltransferasas/inmunología , Bazo/inmunología
3.
Clin Immunol ; 191: 100-109, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29183866

RESUMEN

The proinflammatory protease caspase-1 plays pivotal roles in central pathways of innate immunity, thereby contributing to pathogen clearance. Beside its physiological role, dysregulated activity of caspase-1 is known to contribute to an increasing number of diseases. In this study, we optimized and validated a low-volume human whole blood assay facilitating the measurement of caspase-1 activation and inflammasome-related gene expression upon stimulation of the NLRP3, NLRC4 or AIM2 inflammasome. Using the NLRP3 inflammasome specific inhibitor MCC950, we were able to measure the activity of canonical or alternative NLRP3 pathways, AIM2 and NLRC4 inflammasomes in whole blood. Based on our data we assume a superposition of NLRP3 and NLRC4 inflammasome activities in human whole blood following stimulation with S. typhimurium. The optimized whole blood assay may be suitable for diagnostic and research purposes for pediatric patients who can only donate small amounts of blood.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/sangre , Proteínas de Unión al Calcio/sangre , Proteínas de Unión al ADN/sangre , Inflamasomas/sangre , Proteína con Dominio Pirina 3 de la Familia NLR/sangre , Recolección de Muestras de Sangre , Caspasa 1/fisiología , Humanos , Interleucina-1beta/fisiología , Salmonella typhimurium
4.
Brain Behav Immun ; 65: 202-209, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28495610

RESUMEN

A growing number of studies show an association between seasonal allergic rhinitis (SAR) with depression and anxiety. The underlying mechanisms of a link between SAR and affect, however, are still unclear. The objective of the present study was to investigate depressive symptoms and anxiety in SAR patients and their association to inflammatory and endocrine parameters. SAR patients (n=41) and non-allergic, healthy controls (n=42) were assessed during (pollen season) and out of symptomatic periods (non-pollen season). Inflammatory cytokine profile (Interleukin [IL]-2, IL-4, IL-6, IL-8, IL-10, IL-17, IFN-γ, TNF-α), Immunoglobulin-E (IgE), hair cortisol concentrations (HCC), as well as sleep quality were measured. The present data show that during acute allergic inflammation SAR patients experienced a significant increase in Beck Depression Inventory (BDI-) II scores when (a) compared to the asymptomatic period and (b) when compared to the non-allergic controls, while no differences in anxiety were observed. Increased BDI-II scores in SAR patients were significantly associated with levels of IL-6 as well as IL-6/IL-10 and IFN-γ/IL-10 ratios and further, to an early age at manifestation of SAR and poor sleep quality. These findings support a close relationship between acute allergic processes and affective states, with inflammatory cytokines, sleep, and age of manifestation as potentially relevant mediators.


Asunto(s)
Depresión/inmunología , Rinitis Alérgica Estacional/inmunología , Rinitis Alérgica Estacional/psicología , Adulto , Afecto , Alérgenos/metabolismo , Ansiedad/etiología , Ansiedad/inmunología , Biomarcadores/sangre , Depresión/etiología , Femenino , Humanos , Hipersensibilidad , Inmunoglobulina E/inmunología , Inmunoglobulina E/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Masculino , Polen , Rinitis Alérgica Estacional/metabolismo , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
5.
Klin Padiatr ; 229(3): 113-117, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28561224

RESUMEN

Background Heterozygous point mutations in the GT splice donor consensus sequence of exon 11 of the PIK3R1 gene (coding for p85α, p55α, and p50α regulatory subunits of PI3K) lead to exon skipping and thereby to an aberrant protein that leaves PI3K hyperactivated. Several patients with this particular variant of PI3 kinase delta syndrome (APDS) suffering from sinopulmonary infections and lymphoproliferation have been described. Methods (Whole exome) sequencing, evaluation of cellular and clinical phenotypes. Results We here report a family with a new heterozygous mutation in this gene, a 9 bp deletion (c.1418_1425+1del) that, however, leads to the same skipping of exon 11. The clinical phenotypes of their members partly overlap features of patients of other reports. Conclusions We found a new mutation in PIK3R1 and show how broad the resulting clinical spectrum can be.


Asunto(s)
Variación Genética/genética , Trastornos del Crecimiento/genética , Síndromes de Inmunodeficiencia/genética , Linfoma/genética , Fenotipo , Fosfatidilinositol 3-Quinasas/genética , Mutación Puntual , Linfocitos B/inmunología , Niño , Deleción Cromosómica , Fosfatidilinositol 3-Quinasa Clase Ia , Análisis Mutacional de ADN , Exones/genética , Femenino , Tamización de Portadores Genéticos , Marcadores Genéticos/genética , Trastornos del Crecimiento/diagnóstico , Trastornos del Crecimiento/inmunología , Humanos , Síndromes de Inmunodeficiencia/diagnóstico , Síndromes de Inmunodeficiencia/inmunología , Recuento de Linfocitos , Linfoma/diagnóstico , Linfoma/inmunología , Linaje , Sitios de Empalme de ARN/genética , Secuenciación del Exoma
6.
J Immunol ; 192(9): 4379-85, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24706726

RESUMEN

The proinflammatory enzyme caspase-1 plays an important role in the innate immune system and is involved in a variety of inflammatory conditions. Rare naturally occurring human variants of the caspase-1 gene (CASP1) lead to different protein expression and structure and to decreased or absent enzymatic activity. Paradoxically, a significant number of patients with such variants suffer from febrile episodes despite decreased IL-1ß production and secretion. In this study, we investigate how variant (pro)caspase-1 can possibly contribute to inflammation. In a transfection model, such variant procaspase-1 binds receptor interacting protein kinase 2 (RIP2) via Caspase activation and recruitment domain (CARD)/CARD interaction and thereby activates NF-κB, whereas wild-type procaspase-1 reduces intracellular RIP2 levels by enzymatic cleavage and release into the supernatant. We approach the protein interactions by coimmunoprecipitation and confocal microscopy and show that NF-κB activation is inhibited by anti-RIP2-short hairpin RNA and by the expression of a RIP2 CARD-only protein. In conclusion, variant procaspase-1 binds RIP2 and thereby activates NF-κB. This pathway could possibly contribute to proinflammatory signaling.


Asunto(s)
Caspasa 1/genética , Fiebre/genética , Inflamación/genética , FN-kappa B/metabolismo , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/metabolismo , Western Blotting , Caspasa 1/metabolismo , Fiebre/enzimología , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Variación Genética , Células HEK293 , Humanos , Inmunoprecipitación , Inflamación/inmunología , Inflamación/metabolismo , Transducción de Señal/fisiología , Transducción Genética , Transfección
7.
Ann Rheum Dis ; 74(3): e17, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24445253

RESUMEN

OBJECTIVES: The HIV restriction factor, SAMHD1 (SAM domain and HD domain-containing protein 1), is a triphosphohydrolase that degrades deoxyribonucleoside triphosphates (dNTPs). Mutations in SAMHD1 cause Aicardi-Goutières syndrome (AGS), an inflammatory disorder that shares phenotypic similarity with systemic lupus erythematosus, including activation of antiviral type 1 interferon (IFN). To further define the pathomechanisms underlying autoimmunity in AGS due to SAMHD1 mutations, we investigated the physiological properties of SAMHD1. METHODS: Primary patient fibroblasts were examined for dNTP levels, proliferation, senescence, cell cycle progression and DNA damage. Genome-wide transcriptional profiles were generated by RNA sequencing. Interaction of SAMHD1 with cyclin A was assessed by coimmunoprecipitation and fluorescence cross-correlation spectroscopy. Cell cycle-dependent phosphorylation of SAMHD1 was examined in synchronised HeLa cells and using recombinant SAMHD1. SAMHD1 was knocked down by RNA interference. RESULTS: We show that increased dNTP pools due to SAMHD1 deficiency cause genome instability in fibroblasts of patients with AGS. Constitutive DNA damage signalling is associated with cell cycle delay, cellular senescence, and upregulation of IFN-stimulated genes. SAMHD1 is phosphorylated by cyclin A/cyclin-dependent kinase 1 in a cell cycle-dependent manner, and its level fluctuates during the cell cycle, with the lowest levels observed in G1/S phase. Knockdown of SAMHD1 by RNA interference recapitulates activation of DNA damage signalling and type 1 IFN activation. CONCLUSIONS: SAMHD1 is required for genome integrity by maintaining balanced dNTP pools. dNTP imbalances due to SAMHD1 deficiency cause DNA damage, leading to intrinsic activation of IFN signalling. These findings establish a novel link between DNA damage signalling and innate immune activation in the pathogenesis of autoimmunity.


Asunto(s)
Enfermedades Autoinmunes del Sistema Nervioso/genética , Autoinmunidad/genética , Ciclina A/metabolismo , Fibroblastos/metabolismo , Inestabilidad Genómica/genética , Proteínas de Unión al GTP Monoméricas/genética , Malformaciones del Sistema Nervioso/genética , ARN Mensajero/genética , Enfermedades Autoinmunes del Sistema Nervioso/metabolismo , Proteína Quinasa CDC2 , Células Cultivadas , Quinasas Ciclina-Dependientes/metabolismo , Daño del ADN/genética , Daño del ADN/inmunología , Perfilación de la Expresión Génica , Células HEK293 , Células HeLa , Humanos , Interferón Tipo I/inmunología , Proteínas de Unión al GTP Monoméricas/metabolismo , Malformaciones del Sistema Nervioso/metabolismo , Fosforilación , Interferencia de ARN , Proteína 1 que Contiene Dominios SAM y HD , Transducción de Señal
8.
Curr Rheumatol Rep ; 17(7): 45, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26003867

RESUMEN

Caspase-1 is a proinflammatory enzyme that is essential in many inflammatory conditions including infectious, autoimmune, and autoinflammatory disorders. The inflammation is mainly mediated by the generation of inflammasomes that activate caspase-1 and subsequently interleukin (IL)-1ß and IL-18. In addition, homotypic CARD/CARD interaction of procaspase-1 with RIP2 and thereby activation of the NF-κB pathways may play some role in the inflammation. However, normally, this pathway seems to become downregulated rapidly by the cleavage and excretion of RIP2 by active (pro-)caspase-1. In patients with unexplained recurrent systemic inflammation, CASP1 variants were detected, which often destabilized the caspase-1 dimer interface. Obviously, the resulting decreased or abrogated enzymatic activity and IL-1ß production did not prevent the febrile episodes. As an unexpected finding, the inactive procaspase-1 variants significantly enhanced proinflammatory signaling by increasing RIP2 mediated NF-κB activation in an in vitro cell transfection model. A likely reason is the failure of inactive procaspase-1 to cleave bound RIP2 and also to mediate its excretion out of the intracelluar space thereby keeping the RIP2-NF-κB pathway upregulated. Hence, proinflammatory effects of enzymatically inactive procaspase-1 variants may partially explain the inflammatory episodes of the patients.


Asunto(s)
Enfermedades Autoinmunes/genética , Caspasa 1/genética , Fiebre/genética , Inflamación/genética , Enfermedades Autoinmunes/metabolismo , Caspasa 1/metabolismo , Fiebre/metabolismo , Variación Genética , Humanos , Inmunidad Innata , Inflamación/metabolismo , Recurrencia
9.
Hum Mutat ; 34(1): 122-31, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22833538

RESUMEN

Caspase-1 (Interleukin-1 Converting Enzyme, ICE) is a proinflammatory enzyme that plays pivotal roles in innate immunity and many inflammatory conditions such as periodic fever syndromes and gout. Inflammation is often mediated by enzymatic activation of interleukin (IL)-1ß and IL-18. We detected seven naturally occurring human CASP1 variants with different effects on protein structure, expression, and enzymatic activity. Most mutations destabilized the caspase-1 dimer interface as revealed by crystal structure analysis and homology modeling followed by molecular dynamics simulations. All variants demonstrated decreased or absent enzymatic and IL-1ß releasing activity in vitro, in a cell transfection model, and as low as 25% of normal ex vivo in a whole blood assay of samples taken from subjects with variant CASP1, a subset of whom suffered from unclassified autoinflammation. We conclude that decreased enzymatic activity of caspase-1 is compatible with normal life and does not prevent moderate and severe autoinflammation.


Asunto(s)
Caspasa 1/genética , Caspasa 1/metabolismo , Variación Genética , Interleucina-1beta/metabolismo , Biocatálisis , Caspasa 1/química , Línea Celular , Cristalografía por Rayos X , Citocinas/sangre , Citocinas/metabolismo , Análisis Mutacional de ADN , Predisposición Genética a la Enfermedad/genética , Células HEK293 , Humanos , Inflamación/enzimología , Inflamación/genética , Modelos Moleculares , Mutación , Multimerización de Proteína , Estructura Terciaria de Proteína
10.
Cancer Res ; 83(17): 2858-2872, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37335136

RESUMEN

Genome damage is a main driver of malignant transformation, but it also induces aberrant inflammation via the cGAS/STING DNA-sensing pathway. Activation of cGAS/STING can trigger cell death and senescence, thereby potentially eliminating genome-damaged cells and preventing against malignant transformation. Here, we report that defective ribonucleotide excision repair (RER) in the hematopoietic system caused genome instability with concomitant activation of the cGAS/STING axis and compromised hematopoietic stem cell function, ultimately resulting in leukemogenesis. Additional inactivation of cGAS, STING, or type I IFN signaling, however, had no detectable effect on blood cell generation and leukemia development in RER-deficient hematopoietic cells. In wild-type mice, hematopoiesis under steady-state conditions and in response to genome damage was not affected by loss of cGAS. Together, these data challenge a role of the cGAS/STING pathway in protecting the hematopoietic system against DNA damage and leukemic transformation. SIGNIFICANCE: Loss of cGAS/STING signaling does not impact DNA damage-driven leukemogenesis or alter steady-state, perturbed or malignant hematopoiesis, indicating that the cGAS/STING axis is not a crucial antioncogenic mechanism in the hematopoietic system. See related commentary by Zierhut, p. 2807.


Asunto(s)
Interferón Tipo I , Leucemia , Animales , Ratones , Hematopoyesis/genética , Interferón Tipo I/metabolismo , Leucemia/genética , Nucleotidiltransferasas/genética , Nucleotidiltransferasas/metabolismo , Transducción de Señal
11.
Neurobiol Dis ; 48(3): 339-47, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22782080

RESUMEN

Matrix metalloproteinases (MMPs) are involved in tissue repair, cell death and morphogenesis. We investigated the role of the gelatinases MMP-2 and MMP-9 in the pathogenesis of neuronal death induced by prolonged seizures in the developing brain. Seven-day-old rats, MMP-9 knockout mice and transgenic rats overexpressing MMP-9 received intraperitoneal injections of pilocarpine, 250 mg/kg, to induce seizures. After 6-72 h pups were sacrificed, tissue from different brain regions was isolated and expression of MMP-9 mRNA and protein was analyzed by real-time PCR or Western blot. Additionally, brains were fixed and processed for TUNEL-staining, immunohistochemistry and in situ zymography. We found increased numbers of TUNEL-positive cells 24 h after pilocarpine-induced seizures, most pronounced in cortical areas and the dentate gyrus, and less pronounced in thalamus. At 6-24 h, MMP-9 mRNA levels showed significant elevation compared to sham-treated controls; this effect resolved by 48 h, whereas MMP-2 mRNA levels remained stable. Cortical gelatinolytic activity, monitored by in situ zymography, was enhanced following pilocarpine-induced seizures. The MMP inhibitor GM 6001 ameliorated cell death following pilocarpine-induced seizures in infant rats. MMP-9 knockout mice were less susceptible to seizure-induced brain injury. Transgenic rats overexpressing MMP-9 were equally susceptible to seizure-induced brain injury as wild type rats. Our results suggest a significant contribution of MMP-9 to cell death after pilocarpine-induced seizures in the developing brain. As indicated by Western blot analysis, MMP-9 activation may be linked to activation of the Erk/CREB-pathway. The findings implicate involvement of MMP-9 in the pathophysiology of brain injury following seizures in the developing brain.


Asunto(s)
Apoptosis/fisiología , Encéfalo/enzimología , Metaloproteinasa 9 de la Matriz/metabolismo , Convulsiones/enzimología , Animales , Western Blotting , Encéfalo/patología , Convulsivantes/toxicidad , Humanos , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Metaloproteinasa 9 de la Matriz/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Degeneración Nerviosa , Pilocarpina/toxicidad , ARN Mensajero/análisis , Ratas , Ratas Transgénicas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Convulsiones/inducido químicamente , Convulsiones/patología
12.
Elife ; 112022 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-36314770

RESUMEN

Stimulator of interferon genes (STING) is activated after detection of cytoplasmic dsDNA by cGAS (cyclic GMP-AMP synthase) as part of the innate immunity defence against viral pathogens. STING binds TANK-binding kinase 1 (TBK1). TBK1 mutations are associated with familial amyotrophic lateral sclerosis, and the STING pathway has been implicated in the pathogenesis of further neurodegenerative diseases. To test whether STING activation is sufficient to induce neurodegeneration, we analysed a mouse model that expresses the constitutively active STING variant N153S. In this model, we focused on dopaminergic neurons, which are particularly sensitive to stress and represent a circumscribed population that can be precisely quantified. In adult mice expressing N153S STING, the number of dopaminergic neurons was smaller than in controls, as was the density of dopaminergic axon terminals and the concentration of dopamine in the striatum. We also observed alpha-synuclein pathology and a lower density of synaptic puncta. Neuroinflammation was quantified by staining astroglia and microglia, by measuring mRNAs, proteins and nuclear translocation of transcription factors. These neuroinflammatory markers were already elevated in juvenile mice although at this age the number of dopaminergic neurons was still unaffected, thus preceding the degeneration of dopaminergic neurons. More neuroinflammatory markers were blunted in mice deficient for inflammasomes than in mice deficient for signalling by type I interferons. Neurodegeneration, however, was blunted in both mice. Collectively, these findings demonstrate that chronic activation of the STING pathway is sufficient to cause degeneration of dopaminergic neurons. Targeting the STING pathway could therefore be beneficial in Parkinson's disease and further neurodegenerative diseases.


Neurodegenerative conditions such as Alzheimer's and Parkinson's diseases are characterised by neurons getting damaged and dying. Many factors contribute to this process, but few can be effectively controlled by therapies. Interestingly, previous studies have highlighted that inflammation, a process normally triggered by foreign agents or biological damage, is often associated with neurons degenerating. However, it is unclear whether these responses are the cause or the consequence of brain cell damage. In injured neurons, the genetic information normally contained inside a dedicated cellular compartment can start to leak into the surrounding parts of the cell. This damage triggers an inflammatory response through the STING pathway, a mechanism previously implicated in the onset of Parkinson's disease. In these patients, the neurons that produce the signalling molecule dopamine start to die, leading to difficulty with movement. Whether STING can directly cause this neuronal loss remains unknown. To answer this question, Szegö, Malz et al. genetically engineered mice in which the STING pathway is permanently activated. The animals had fewer dopamine-producing neurons and accumulated harmful clumps of proteins; both these biological features are characteristic signs of Parkinson's disease. Crucially, signs of inflammation were present before neurons started to show damage, suggesting that inflammatory responses could cause neurodegeneration. Further experiments revealed that STING triggers several molecular cascades; blocking one only of these pathways did not keep the neurons healthy. Neurodegenerative diseases are a growing concern around the world. The results from Szegö, Malz et al. suggest that preventing prolonged inflammatory may reduce the risk of neurodegeneration. If further research confirms these findings, in particular in humans, well-known treatments against inflammation could potentially become relevant to fight these conditions.


Asunto(s)
Neuronas Dopaminérgicas , Enfermedades Neuroinflamatorias , Animales , Ratones , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Microglía/metabolismo , Enfermedades Neurodegenerativas/patología , Enfermedades Neuroinflamatorias/genética , Enfermedades Neuroinflamatorias/metabolismo , Enfermedad de Parkinson/genética
13.
STAR Protoc ; 3(2): 101309, 2022 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-35496781

RESUMEN

Activation of innate immune sensors by endogenous DNA and RNA can lead to autoimmune and autoinflammatory diseases. Quantification of the unperturbed phosphoprotein content in immune cells provides insight into the spontaneous activity of immune signaling pathways triggered by nucleic acid recognition. Here, we present a phosphoflow protocol for measuring phosphoproteins in mouse models of autoimmunity that incorporates strategies to preserve native phosphoprotein levels during sample collection and to reliably detect low signaling activity common in chronic disease states. For complete details on the use and execution of this protocol, please refer to Jütte et al. (2021).


Asunto(s)
Enfermedades Autoinmunes , Ácidos Nucleicos , Animales , Enfermedades Autoinmunes/genética , Autoinmunidad , Ratones , Fosfoproteínas , Transducción de Señal/fisiología
14.
J Exp Med ; 219(10)2022 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-35997679

RESUMEN

Autoimmune vasculitis is a group of life-threatening diseases, whose underlying pathogenic mechanisms are incompletely understood, hampering development of targeted therapies. Here, we demonstrate that patients suffering from anti-neutrophil cytoplasmic antibodies (ANCA)-associated vasculitis (AAV) showed increased levels of cGAMP and enhanced IFN-I signature. To identify disease mechanisms and potential therapeutic targets, we developed a mouse model for pulmonary AAV that mimics severe disease in patients. Immunogenic DNA accumulated during disease onset, triggering cGAS/STING/IRF3-dependent IFN-I release that promoted endothelial damage, pulmonary hemorrhages, and lung dysfunction. Macrophage subsets played dichotomic roles in disease. While recruited monocyte-derived macrophages were major disease drivers by producing most IFN-ß, resident alveolar macrophages contributed to tissue homeostasis by clearing red blood cells and limiting infiltration of IFN-ß-producing macrophages. Moreover, pharmacological inhibition of STING, IFNAR-I, or its downstream JAK/STAT signaling reduced disease severity and accelerated recovery. Our study unveils the importance of STING/IFN-I axis in promoting pulmonary AAV progression and identifies cellular and molecular targets to ameliorate disease outcomes.


Asunto(s)
Interferón Tipo I , Ácidos Nucleicos , Vasculitis , Animales , Pulmón , Macrófagos , Proteínas de la Membrana/metabolismo , Ratones , Nucleotidiltransferasas
15.
Cell Rep ; 30(8): 2501-2511.e5, 2020 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-32101731

RESUMEN

Pro-inflammatory caspase-1 is a key player in innate immunity. Caspase-1 processes interleukin (IL)-1ß and IL-18 to their mature forms and triggers pyroptosis. These caspase-1 functions are linked to its enzymatic activity. However, loss-of-function missense mutations in CASP1 do not prevent autoinflammation in patients, despite decreased IL-1ß production. In vitro data suggest that enzymatically inactive caspase-1 drives inflammation via enhanced nuclear factor κB (NF-κB) activation, independent of IL-1ß processing. Here, we report two mouse models of enzymatically inactive caspase-1-C284A, demonstrating the relevance of this pathway in vivo. In contrast to Casp1-/- mice, caspase-1-C284A mice show pronounced hypothermia and increased levels of the pro-inflammatory cytokines tumor necrosis factor alpha (TNF-α) and IL-6 when challenged with lipopolysaccharide (LPS). Caspase-1-C284A signaling is RIP2 dependent and mediated by TNF-α but independent of the NLRP3 inflammasome. LPS-stimulated whole blood from patients carrying loss-of-function missense mutations in CASP1 secretes higher amounts of TNF-α. Taken together, these results reveal non-canonical caspase-1 signaling in vivo.


Asunto(s)
Caspasa 1/metabolismo , Inflamación/patología , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo , Adolescente , Adulto , Animales , Niño , Preescolar , Sitios Genéticos , Genotipo , Células HEK293 , Heterocigoto , Humanos , Ratones Endogámicos C57BL , Mutación/genética , Adulto Joven
16.
Cell Rep ; 31(11): 107771, 2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32553167

RESUMEN

STING gain-of-function causes autoimmunity and immunodeficiency in mice and STING-associated vasculopathy with onset in infancy (SAVI) in humans. Here, we report that STING gain-of-function in mice prevents development of lymph nodes and Peyer's patches. We show that the absence of secondary lymphoid organs is associated with diminished numbers of innate lymphoid cells (ILCs), including lymphoid tissue inducer (LTi) cells. Although wild-type (WT) α4ß7+ progenitors differentiate efficiently into LTi cells, STING gain-of-function progenitors do not. Furthermore, STING gain-of-function impairs development of all types of ILCs. Patients with STING gain-of-function mutations have fewer ILCs, although they still have lymph nodes. In mice, expression of the STING mutant in RORγT-positive lineages prevents development of lymph nodes and reduces numbers of LTi cells. RORγT lineage-specific expression of STING gain-of-function also causes lung disease. Since RORγT is expressed exclusively in LTi cells during fetal development, our findings suggest that STING gain-of-function prevents lymph node organogenesis by reducing LTi cell numbers in mice.


Asunto(s)
Diferenciación Celular/inmunología , Inmunidad Innata/inmunología , Ganglios Linfáticos/inmunología , Linfocitos/citología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Mutación con Ganancia de Función/inmunología , Tejido Linfoide/inmunología , Ratones , Organogénesis/inmunología
17.
Histochem Cell Biol ; 132(4): 435-45, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19526364

RESUMEN

Emerging evidence suggests a role for glutamate and its receptors in the biology of cancer. This study was designed to systematically analyze the expression of ionotropic and metabotropic glutamate receptor subunits in various human cancer cell lines, compare expression levels to those in human brain tissue and, using electrophysiological techniques, explore whether cancer cells respond to glutamate receptor agonists and antagonists. Expression analysis of glutamate receptor subunits NR1-NR3B, GluR1-GluR7, KA1, KA2 and mGluR1-mGluR8 was performed by means of RT-PCR in human rhabdomyosarcoma/medulloblastoma (TE671), neuroblastoma (SK-NA-S), thyroid carcinoma (FTC 238), lung carcinoma (SK-LU-1), astrocytoma (MOGGCCM), multiple myeloma (RPMI 8226), glioma (U87-MG and U343), lung carcinoma (A549), colon adenocarcinoma (HT 29), T cell leukemia cells (Jurkat E6.1), breast carcinoma (T47D) and colon adenocarcinoma (LS180). Analysis revealed that all glutamate receptor subunits were differentially expressed in the tumor cell lines. For the majority of tumors, expression levels of NR2B, GluR4, GluR6 and KA2 were lower compared to human brain tissue. Confocal imaging revealed that selected glutamate receptor subunit proteins were expressed in tumor cells. By means of patch-clamp analysis, it was shown that A549 and TE671 cells depolarized in response to application of glutamate agonists and that this effect was reversed by glutamate receptor antagonists. This study reveals that glutamate receptor subunits are differentially expressed in human tumor cell lines at the mRNA and the protein level, and that their expression is associated with the formation of functional channels. The potential role of glutamate receptor antagonists in cancer therapy is a feasible goal to be explored in clinical trials.


Asunto(s)
Neoplasias/metabolismo , Receptores AMPA/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , 6-Ciano 7-nitroquinoxalina 2,3-diona/farmacología , Encéfalo/metabolismo , Línea Celular Tumoral , Antagonistas de Aminoácidos Excitadores/farmacología , Humanos , Técnicas de Placa-Clamp , Subunidades de Proteína/efectos de los fármacos , Subunidades de Proteína/metabolismo , Receptores AMPA/antagonistas & inhibidores , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Valina/análogos & derivados , Valina/farmacología
18.
Anticancer Agents Med Chem ; 18(4): 591-596, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29493464

RESUMEN

BACKGROUND: Glutamate receptors are widely expressed in different types of cancer cells. α-Amino-3- hydroxy-5-methyl-4-isoxazolepropionate (AMPA) receptors are ionotropic glutamate receptors which are coupled to intracellular signaling pathways that influence cancer cell survival, proliferation, and migration. Blockade of AMPA receptors by pharmacologic compounds may potentially constitute an effective tool in anticancer treatment strategies. METHOD: Here we investigated the impact of the AMPA receptor antagonist CFM-2 on the expression of the protein survivin, which is known to promote cancer cell survival and proliferation. We show that CFM-2 inhibits survivin expression at mRNA and protein levels and decreases the viability of cancer cells. Using a stably transfected cell line which overexpresses survivin, we demonstrate that over-expression of survivin enhances cancer cell viability and attenuates CFM-2-mediated inhibition of cancer cell growth. RESULT: These findings point towards suppression of survivin expression as a new mechanism contributing to anticancer effects of AMPA antagonists.


Asunto(s)
Antineoplásicos/farmacología , Benzodiazepinonas/farmacología , Receptores AMPA/antagonistas & inhibidores , Células A549 , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Estructura Molecular , Receptores AMPA/metabolismo , Relación Estructura-Actividad , Células Tumorales Cultivadas
20.
Pediatr Rheumatol Online J ; 14(1): 63, 2016 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-27881174

RESUMEN

BACKGROUND: The pathophysiological origin of juvenile idiopathic arthritis (JIA) is largely unknown. However, individuals with presumably pathogenic mutations in FAMIN have been reported, associating this gene with a rare subtype of this disorder. FAMIN, that is formerly also referred to as LACC1 or C13orf31, has recently been shown to play a crucial role in immune-metabolic functions and is involved in regulation of inflammasome activation and promotion of ROS production. CASE PRESENTATION: We describe two siblings with severe familial forms of juvenile arthritis in which whole-exome-sequencing revealed a novel homozygous frameshift mutation (NM_153218.2:c.827delC¸. p.(T276fs*2) in FAMIN. CONCLUSIONS: The observation of a new deleterious mutation adds further evidence that pathogenic mutations in FAMIN are causal for a monogenic form of JIA. Furthermore the associated phenotype is not restricted to systemic JIA, but can also be found in other forms of familial juvenile arthritis.


Asunto(s)
Artritis Juvenil/genética , Mutación del Sistema de Lectura/genética , Proteínas/genética , Niño , Exoma/genética , Femenino , Predisposición Genética a la Enfermedad/genética , Homocigoto , Humanos , Péptidos y Proteínas de Señalización Intracelular , Linaje , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA