Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
1.
ACS Nano ; 12(12): 11949-11962, 2018 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-30444603

RESUMEN

Graphene oxide (GO), an oxidized form of graphene, has potential applications in biomedical research. However, how GO interacts with biological systems, including the innate immune system, is poorly understood. Here, we elucidate the effects of GO sheets on macrophages, identifying distinctive effects of GO on the inflammatory phenotype. Small, thin (s)-GO dose-dependently inhibited release of interleukin (IL)-1ß and IL-6 but not tumor necrosis factor α. NLRP3 inflammasome and caspase-1 activation was not affected. The effect of s-GO was pretranslational, as s-GO blocked Toll-like receptor 4-dependent expression of Il1b and Il6 but not Nlrp3 or Tnf mRNA transcripts. s-GO was internalized by immortalized bone-marrow-derived macrophages, suggesting a potential intracellular action. Uptake of polystyrene beads with similar lateral dimensions and surface charge did not phenocopy the effects of s-GO, suggesting that s-GO-mediated inhibition of interleukin expression was not simply due to particle phagocytosis. RNA-Seq analysis established that s-GO had profound effects on the immunometabolism of the cells, leading to activation of the transcription factor nuclear factor erythroid 2-related factor 2, which inhibited expression of cytokines such as IL-1ß and IL-6. Thus, we have identified immunometabolic effects of GO that reveal another dimension to its effects on cells. These findings suggest that s-GO may be used as a valuable tool to generate further insights into inflammatory mechanisms and indicate its potential applications in biomedicine.


Asunto(s)
Citocinas/metabolismo , Grafito/química , Factor 2 Relacionado con NF-E2/metabolismo , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Inflamasomas/inmunología , Inflamasomas/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Neuroglía/metabolismo , ARN Mensajero/metabolismo , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
2.
Stroke ; 49(5): 1210-1216, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29567761

RESUMEN

BACKGROUND AND PURPOSE: The proinflammatory cytokine IL-1 (interleukin-1) has a deleterious role in cerebral ischemia, which is attenuated by IL-1 receptor antagonist (IL-1Ra). IL-1 induces peripheral inflammatory mediators, such as interleukin-6, which are associated with worse prognosis after ischemic stroke. We investigated whether subcutaneous IL-1Ra reduces the peripheral inflammatory response in acute ischemic stroke. METHODS: SCIL-STROKE (Subcutaneous Interleukin-1 Receptor Antagonist in Ischemic Stroke) was a single-center, double-blind, randomized, placebo-controlled phase 2 trial of subcutaneous IL-1Ra (100 mg administered twice daily for 3 days) in patients presenting within 5 hours of ischemic stroke onset. Randomization was stratified for baseline National Institutes of Health Stroke Scale score and thrombolysis. Measurement of plasma interleukin-6 and other peripheral inflammatory markers was undertaken at 5 time points. The primary outcome was difference in concentration of log(interleukin-6) as area under the curve to day 3. Secondary outcomes included exploratory effect of IL-1Ra on 3-month outcome with the modified Rankin Scale. RESULTS: We recruited 80 patients (mean age, 72 years; median National Institutes of Health Stroke Scale, 12) of whom 73% received intravenous thrombolysis with alteplase. IL-1Ra significantly reduced plasma interleukin-6 (P<0.001) and plasma C-reactive protein (P<0.001). IL-1Ra was well tolerated with no safety concerns. Allocation to IL-1Ra was not associated with a favorable outcome on modified Rankin Scale: odds ratio (95% confidence interval)=0.67 (0.29-1.52), P=0.34. Exploratory mediation analysis suggested that IL-1Ra improved clinical outcome by reducing inflammation, but there was a statistically significant, alternative mechanism countering this benefit. CONCLUSIONS: IL-1Ra reduced plasma inflammatory markers which are known to be associated with worse clinical outcome in ischemic stroke. Subcutaneous IL-1Ra is safe and well tolerated. Further experimental studies are required to investigate efficacy and possible interactions of IL-1Ra with thrombolysis. CLINICAL TRIAL REGISTRATION: URL: http://www.clinicaltrials.gov. Unique identifier: ISRCTN74236229.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Fibrinolíticos/uso terapéutico , Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico , Accidente Cerebrovascular/tratamiento farmacológico , Activador de Tejido Plasminógeno/uso terapéutico , Anciano , Anciano de 80 o más Años , Área Bajo la Curva , Isquemia Encefálica/inmunología , Proteína C-Reactiva/inmunología , Método Doble Ciego , Femenino , Humanos , Inflamación , Inyecciones Subcutáneas , Interleucina-6/inmunología , Masculino , Persona de Mediana Edad , Oportunidad Relativa , Accidente Cerebrovascular/inmunología , Terapia Trombolítica , Resultado del Tratamiento
3.
Cell Chem Biol ; 24(11): 1321-1335.e5, 2017 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-28943355

RESUMEN

NLRP3 is a receptor important for host responses to infection, yet is also known to contribute to devastating diseases such as Alzheimer's disease, diabetes, atherosclerosis, and others, making inhibitors for NLRP3 sought after. One of the inhibitors currently in use is 2-aminoethoxy diphenylborinate (2APB). Unfortunately, in addition to inhibiting NLRP3, 2APB also displays non-selective effects on cellular Ca2+ homeostasis. Here, we use 2APB as a chemical scaffold to build a series of inhibitors, the NBC series, which inhibit the NLRP3 inflammasome in vitro and in vivo without affecting Ca2+ homeostasis. The core chemical insight of this work is that the oxazaborine ring is a critical feature of the NBC series, and the main biological insight the use of NBC inhibitors led to was that NLRP3 inflammasome activation was independent of Ca2+. The NBC compounds represent useful tools to dissect NLRP3 function, and may lead to oxazaborine ring-containing therapeutics.


Asunto(s)
Boro/química , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Animales , Células de la Médula Ósea/citología , Boro/farmacología , Compuestos de Boro/química , Compuestos de Boro/metabolismo , Compuestos de Boro/farmacología , Calcio/metabolismo , Células Cultivadas , Cristalografía por Rayos X , Humanos , Concentración 50 Inhibidora , Interleucina-1beta/metabolismo , Lipopolisacáridos/toxicidad , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Conformación Molecular , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Relación Estructura-Actividad
4.
Stem Cell Res Ther ; 8(1): 79, 2017 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-28412968

RESUMEN

BACKGROUND: Inflammation is a key contributor to central nervous system (CNS) injury such as stroke, and is a major target for therapeutic intervention. Effective treatments for CNS injuries are limited and applicable to only a minority of patients. Stem cell-based therapies are increasingly considered for the treatment of CNS disease, because they can be used as in-situ regulators of inflammation, and improve tissue repair and recovery. One promising option is the use of bone marrow-derived mesenchymal stem cells (MSCs), which can secrete anti-inflammatory and trophic factors, can migrate towards inflamed and injured sites or can be implanted locally. Here we tested the hypothesis that pre-treatment with inflammatory cytokines can prime MSCs towards an anti-inflammatory and pro-trophic phenotype in vitro. METHODS: Human MSCs from three different donors were cultured in vitro and treated with inflammatory mediators as follows: interleukin (IL)-1α, IL-1ß, tumour necrosis factor alpha (TNF-α) or interferon-γ. After 24 h of treatment, cell supernatants were analysed by ELISA for expression of granulocyte-colony stimulating factor (G-CSF), IL-10, brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), IL-1 receptor antagonist (IL-1Ra) and vascular endothelial growth factor (VEGF). To confirm the anti-inflammatory potential of MSCs, immortalised mouse microglial BV2 cells were treated with bacterial lipopolysaccharide (LPS) and exposed to conditioned media (CM) of naïve or IL-1-primed MSCs, and levels of secreted microglial-derived inflammatory mediators including TNF-α, IL-10, G-CSF and IL-6 were measured by ELISA. RESULTS: Unstimulated MSCs constitutively expressed anti-inflammatory cytokines and trophic factors (IL-10, VEGF, BDNF, G-CSF, NGF and IL-1Ra). MSCs primed with IL-1α or IL-1ß showed increased secretion of G-CSF, which was blocked by IL-1Ra. Furthermore, LPS-treated BV2 cells secreted less inflammatory and apoptotic markers, and showed increased secretion of the anti-inflammatory IL-10 in response to treatment with CM of IL-1-primed MSCs compared with CM of unprimed MSCs. CONCLUSIONS: Our results demonstrate that priming MSCs with IL-1 increases expression of trophic factor G-CSF through an IL-1 receptor type 1 (IL-1R1) mechanism, and induces a reduction in the secretion of inflammatory mediators in LPS-activated microglial cells. The results therefore support the potential use of preconditioning treatments of stem cells in future therapies.


Asunto(s)
Antiinflamatorios/farmacología , Interleucina-1alfa/farmacología , Interleucina-1beta/farmacología , Células Madre Mesenquimatosas/citología , Adulto , Animales , Biomarcadores/metabolismo , Medios de Cultivo Condicionados/farmacología , Femenino , Factor Estimulante de Colonias de Granulocitos/farmacología , Humanos , Interleucina-6/metabolismo , Lipopolisacáridos/farmacología , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Microglía/citología , Microglía/efectos de los fármacos , Factores de Crecimiento Nervioso/farmacología , Fenotipo , Factor de Necrosis Tumoral alfa/metabolismo , Adulto Joven
5.
Brain Behav Immun ; 61: 117-126, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27856349

RESUMEN

Neuroprotective strategies for ischemic stroke have failed to translate from bench to bedside, possibly due to the lack of consideration of key clinical co-morbidities. Stroke and co-morbidities are associated with raised levels of the pro-inflammatory cytokine interleukin-1 (IL-1). Inhibition of IL-1 by the administration of interleukin-1 receptor antagonist (IL-1Ra) has shown to be neuroprotective after experimental cerebral ischemia. Stroke can also trigger a robust neuroreparative response following injury, yet many of these new born neurons fail to survive or integrate into pre-existing circuits. Thus, we explore here effects of IL-1Ra on post-stroke neurogenesis in young and aged/co-morbid rats. Aged lean, aged Corpulent (a model of atherosclerosis, obesity and insulin resistance) and young Wistar male rats were exposed to transient cerebral ischemia, received subcutaneous IL-1Ra 3 and 6h during reperfusion, and effects on stroke outcome and neurogenesis were analyzed. Our results show that administration of IL-1Ra improves stroke outcome in both young and aged/co-morbid rats. Furthermore, IL-1Ra not only increases stem cell proliferation, but also significantly enhances neuroblast migration and the number of newly born neurons after cerebral ischemia. Overall, our data demonstrate that systemic administration of IL-1Ra improves outcome and promotes neurogenesis after experimental stroke, further highlighting the therapeutic potential of this clinically approved drug.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico , Neurogénesis/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Movimiento Celular/efectos de los fármacos , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Masculino , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Ratas , Ratas Wistar , Factores de Tiempo
6.
Stroke ; 47(5): 1312-1318, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27032444

RESUMEN

BACKGROUND AND PURPOSE: The debate over the fact that experimental drugs proposed for the treatment of stroke fail in the translation to the clinical situation has attracted considerable attention in the literature. In this context, we present a retrospective pooled analysis of a large data set from preclinical studies, to examine the effects of early versus late administration of intravenous recombinant tissue-type plasminogen activator. METHODS: We collected data from 26 individual studies from 9 international centers (13 researchers; 716 animals) that compared recombinant tissue-type plasminogen activator with controls, in a unique mouse model of thromboembolic stroke induced by an in situ injection of thrombin into the middle cerebral artery. Studies were classified into early (<3 hours) versus late (≥3 hours) drug administration. Final infarct volumes, assessed by histology or magnetic resonance imaging, were compared in each study, and the absolute differences were pooled in a random-effect meta-analysis. The influence of time of administration was tested. RESULTS: When compared with saline controls, early recombinant tissue-type plasminogen activator administration was associated with a significant benefit (absolute difference, -6.63 mm(3); 95% confidence interval, -9.08 to -4.17; I(2)=76%), whereas late recombinant tissue-type plasminogen activator treatment showed a deleterious effect (+5.06 mm(3); 95% confidence interval, +2.78 to +7.34; I(2)=42%; Pint<0.00001). Results remained unchanged after subgroup analyses. CONCLUSIONS: Our results provide the basis needed for the design of future preclinical studies on recanalization therapies using this model of thromboembolic stroke in mice. The power analysis reveals that a multicenter trial would require 123 animals per group instead of 40 for a single-center trial.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Fibrinolíticos/farmacología , Accidente Cerebrovascular/tratamiento farmacológico , Activador de Tejido Plasminógeno/farmacología , Animales , Isquemia Encefálica/patología , Modelos Animales de Enfermedad , Fibrinolíticos/administración & dosificación , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Accidente Cerebrovascular/patología , Activador de Tejido Plasminógeno/administración & dosificación
8.
J Cereb Blood Flow Metab ; 36(3): 596-605, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26661169

RESUMEN

Stroke represents a global challenge and is a leading cause of permanent disability worldwide. Despite much effort, translation of research findings to clinical benefit has not yet been successful. Failure of neuroprotection trials is considered, in part, due to the low quality of preclinical studies, low level of reproducibility across different laboratories and that stroke co-morbidities have not been fully considered in experimental models. More rigorous testing of new drug candidates in different experimental models of stroke and initiation of preclinical cross-laboratory studies have been suggested as ways to improve translation. However, to our knowledge, no drugs currently in clinical stroke trials have been investigated in preclinical cross-laboratory studies. The cytokine interleukin 1 is a key mediator of neuronal injury, and the naturally occurring interleukin 1 receptor antagonist has been reported as beneficial in experimental studies of stroke. In the present paper, we report on a preclinical cross-laboratory stroke trial designed to investigate the efficacy of interleukin 1 receptor antagonist in different research laboratories across Europe. Our results strongly support the therapeutic potential of interleukin 1 receptor antagonist in experimental stroke and provide further evidence that interleukin 1 receptor antagonist should be evaluated in more extensive clinical stroke trials.


Asunto(s)
Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Receptores de Interleucina-1/antagonistas & inhibidores , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Encéfalo/patología , Edema Encefálico/complicaciones , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/inmunología , Edema Encefálico/patología , Isquemia Encefálica/complicaciones , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/inmunología , Isquemia Encefálica/patología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Inflamación/complicaciones , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Inflamación/patología , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Receptores de Interleucina-1/inmunología , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/inmunología , Accidente Cerebrovascular/patología
9.
Eur J Immunol ; 46(4): 912-8, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26692072

RESUMEN

IL-1 is a key cytokine known to drive chronic inflammation and to regulate many physiological, immunological, and neuroimmunological responses via actions on diverse cell types of the body. To determine the mechanisms of IL-1 actions as part of the inflammatory response in vivo, we generated a conditional IL-1 receptor 1 (IL-1R1) mouse mutant using the Cre/LoxP system (IL-1R1(fl/fl) ). In the mutant generated, exon 5, which encodes part of the extracellular-binding region of the receptor, is flanked by LoxP sites, thereby inactivating the two previously described functional IL-1R1 gene transcripts after Cre-mediated recombination. Using keratin 14-Cre driver mice, new IL-1R1 deficient (-/-) mice were subsequently generated, in which all signaling IL-1 receptor isoforms are deleted ubiquitously. Furthermore, using vav-iCre driver mice, we deleted IL-1 receptor isoforms in the hematopoietic system. In these mice, we show that both the IL-17 and IL-22 cytokine response is reduced, when mice are challenged by the helminth Trichuris muris. We are currently crossing IL-1R1(fl/fl) mice with different Cre-expressing mice in order to study mechanisms of acute and chronic inflammatory diseases.


Asunto(s)
Inflamación/inmunología , Interleucina-17/biosíntesis , Interleucinas/biosíntesis , Receptores Tipo I de Interleucina-1/genética , Trichuris/inmunología , Animales , Interleucina-17/inmunología , Interleucinas/inmunología , Queratina-14/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Tipo I de Interleucina-1/inmunología , Interleucina-22
10.
Exp Physiol ; 100(12): 1488-94, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26096539

RESUMEN

NEW FINDINGS: What is the topic of this review? This review discusses the latest findings on the contribution of inflammation to brain injury, how inflammation is a therapeutic target, and details of recent and forthcoming clinical studies. What advances does it highlight? Here we highlight recent advances on the role and regulation of inflammasomes, and the latest clinical progress in targeting inflammation. Acute brain injury is one of the leading causes of mortality and disability worldwide. Despite this, treatments for acute brain injuries are limited, and there remains a massive unmet clinical need. Inflammation has emerged as a major contributor to non-communicable diseases, and there is now substantial and growing evidence that inflammation, driven by the cytokine interleukin-1 (IL-1), worsens acute brain injury. Interleukin-1 is regulated by large, multimolecular complexes called inflammasomes. Here, we discuss the latest research on the regulation of inflammasomes and IL-1 in the brain, preclinical efforts to establish the IL-1 system as a therapeutic target, and the promise of recent and future clinical studies on blocking the action of IL-1 for the treatment of brain injury.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Interleucina-1/farmacología , Interleucina-1/uso terapéutico , Animales , Humanos , Inflamasomas/efectos de los fármacos , Inflamación/tratamiento farmacológico
11.
Eur J Immunol ; 45(2): 525-30, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25367678

RESUMEN

The immune system is implicated in a wide range of disorders affecting the brain and is, therefore, an attractive target for therapy. Interleukin-1 (IL-1) is a potent regulator of the innate immune system important for host defense but is also associated with injury and disease in the brain. Here, we show that IL-1 is a key mediator driving an innate immune response to inflammatory challenge in the mouse brain but is dispensable in extracerebral tissues including the lung and peritoneum. We also demonstrate that IL-1α is an important ligand contributing to the CNS dependence on IL-1 and that IL-1 derived from the CNS compartment (most likely microglia) is the major source driving this effect. These data reveal previously unknown tissue-specific requirements for IL-1 in driving innate immunity and suggest that IL-1-mediated inflammation in the brain could be selectively targeted without compromising systemic innate immune responses that are important for resistance to infection. This property could be exploited to mitigate injury- and disease-associated inflammation in the brain without increasing susceptibility to systemic infection, an important complication in several neurological disorders.


Asunto(s)
Encéfalo/inmunología , Encefalitis/inmunología , Interleucina-1alfa/genética , Interleucina-1beta/genética , Microglía/inmunología , Transducción de Señal/inmunología , Animales , Encéfalo/patología , Encefalitis/inducido químicamente , Encefalitis/genética , Encefalitis/patología , Regulación de la Expresión Génica , Inmunidad Innata , Inyecciones Intraventriculares , Interleucina-1alfa/deficiencia , Interleucina-1alfa/inmunología , Interleucina-1beta/deficiencia , Interleucina-1beta/inmunología , Lipopolisacáridos , Pulmón/inmunología , Ratones , Ratones Noqueados , Microglía/patología , Infiltración Neutrófila , Neutrófilos/inmunología , Neutrófilos/patología , Especificidad de Órganos , Peritoneo/inmunología
12.
Behav Brain Res ; 270: 18-28, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24821402

RESUMEN

Cerebral ischemia is one of the most common causes of disabilities in adults and leads to long-term motor and cognitive impairments with limited therapeutic possibilities. Treatment options have proven efficient in preclinical models of cerebral ischemia but have failed in the clinical setting. This limited translation may be due to the suitability of models used and outcomes measured as most studies have focused on the early period after injury with gross motor scales, which have limited correlation to the clinical situation. The aim of this study was to determine long-term functional outcomes after cerebral ischemia in rats, focusing on fine motor function, social and depressive behavior as clinically relevant measures. A secondary objective was to evaluate the effects of an anti-inflammatory treatment (interleukin-1 receptor antagonist (IL-1Ra)) on functional recovery and compensation. Infarct volume was correlated with long-term (25 days) impairments in fine motor skills, but not with emotional components of behavior. Motor impairments could not be detected using conventional neurological tests and only detailed analysis allowed differentiation between recovery and compensation. Acute systemic administration of IL-1Ra (at reperfusion) led to a faster and more complete recovery, but delayed (24h) IL-1Ra treatment had no effect. In summary functional assessment after brain injury requires detailed motor tests in order to address long-term impairments and compensation processes that are mediated by intact tissues. Functional deficits in skilled movement after brain injury represent ideal predictors of long-term outcomes and should become standard measures in the assessment of preclinical animal models.


Asunto(s)
Isquemia Encefálica/fisiopatología , Isquemia Encefálica/psicología , Encéfalo/patología , Actividad Motora , Conducta Social , Animales , Antirreumáticos/administración & dosificación , Encéfalo/efectos de los fármacos , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/patología , Depresión/tratamiento farmacológico , Depresión/psicología , Modelos Animales de Enfermedad , Proteína Antagonista del Receptor de Interleucina 1/administración & dosificación , Masculino , Actividad Motora/efectos de los fármacos , Ratas , Ratas Wistar , Recuperación de la Función , Factores de Tiempo , Resultado del Tratamiento
13.
J Biol Chem ; 289(23): 15942-50, 2014 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-24790078

RESUMEN

The cytokine interleukin-1 (IL-1) has two main pro-inflammatory forms, IL-1α and IL-1ß, which are central to host responses to infection and to damaging sterile inflammation. Processing of IL-1 precursor proteins to active cytokines commonly occurs through activation of proteases, notably caspases and calpains. These proteases are instrumental in cell death, and inflammation and cell death are closely associated, hence we sought to determine the impact of cell death pathways on IL-1 processing and release. We discovered that apoptotic regulation of caspase-8 specifically induced the processing and release of IL-1ß. Conversely, necroptosis caused the processing and release of IL-1α, and this was independent of IL-1ß processing and release. These data suggest that the mechanism through which an IL-1-expressing cell dies dictates the nature of the inflammatory mechanism that follows. These insights may allow modification of inflammation through the selective targeting of cell death mechanisms during disease.


Asunto(s)
Apoptosis , Interleucina-1alfa/metabolismo , Interleucina-1beta/metabolismo , Animales , Calcio/metabolismo , Calpaína/metabolismo , Caspasa 8/metabolismo , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Necrosis
14.
Ann Neurol ; 75(5): 670-83, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24644058

RESUMEN

OBJECTIVE: Bacterial infection contributes to diverse noninfectious diseases and worsens outcome after stroke. Streptococcus pneumoniae, the most common infection in patients at risk of stroke, is a major cause of prolonged hospitalization and death of stroke patients, but how infection impacts clinical outcome is not known. METHODS: We induced sustained pulmonary infection by a human S. pneumoniae isolate in naive and comorbid rodents to investigate the effect of infection on vascular and inflammatory responses prior to and after cerebral ischemia. RESULTS: S. pneumoniae infection triggered atherogenesis, led to systemic induction of interleukin (IL) 1, and profoundly exacerbated (50-90%) ischemic brain injury in rats and mice, a response that was more severe in combination with old age and atherosclerosis. Systemic blockade of IL-1 with IL-1 receptor antagonist (IL-1Ra) fully reversed infection-induced exacerbation of brain injury and functional impairment caused by cerebral ischemia. We show that infection-induced systemic inflammation mediates its effects via increasing platelet activation and microvascular coagulation in the brain after cerebral ischemia, as confirmed by reduced brain injury in response to blockade of platelet glycoprotein (GP) Ibα. IL-1 and platelet-mediated signals converge on microglia, as both IL-1Ra and GPIbα blockade reversed the production of IL-1α by microglia in response to cerebral ischemia in infected animals. INTERPRETATION: S. pneumoniae infection augments atherosclerosis and exacerbates ischemic brain injury via IL-1 and platelet-mediated systemic inflammation. These mechanisms may contribute to diverse cardio- and cerebrovascular pathologies in humans.


Asunto(s)
Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Interleucina-1/efectos adversos , Complejo GPIb-IX de Glicoproteína Plaquetaria/efectos adversos , Infecciones Estreptocócicas/metabolismo , Infecciones Estreptocócicas/patología , Streptococcus pneumoniae , Animales , Isquemia Encefálica/microbiología , Progresión de la Enfermedad , Humanos , Inflamación/metabolismo , Inflamación/microbiología , Inflamación/patología , Interleucina-1/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Microglía/microbiología , Microglía/patología , Activación Plaquetaria , Complejo GPIb-IX de Glicoproteína Plaquetaria/antagonistas & inhibidores , Complejo GPIb-IX de Glicoproteína Plaquetaria/fisiología , Ratas , Ratas Wistar , Infecciones Estreptocócicas/microbiología
15.
J Neuroinflammation ; 11: 1, 2014 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-24383930

RESUMEN

BACKGROUND: Interleukin-1 (IL-1) is a key mediator of ischaemic brain injury induced by stroke and subarachnoid haemorrhage (SAH). IL-1 receptor antagonist (IL-1Ra) limits brain injury in experimental stroke and reduces plasma inflammatory mediators associated with poor outcome in ischaemic stroke patients. Intravenous (IV) IL-1Ra crosses the blood-brain barrier (BBB) in patients with SAH, to achieve cerebrospinal fluid (CSF) concentrations that are neuroprotective in rats. METHODS: A small phase II, double-blind, randomised controlled study was carried out across two UK neurosurgical centres with the aim of recruiting 32 patients. Adult patients with aneurysmal SAH, requiring external ventricular drainage (EVD) within 72 hours of ictus, were eligible. Patients were randomised to receive IL-1Ra (500 mg bolus, then a 10 mg/kg/hr infusion for 24 hours) or placebo. Serial samples of CSF and plasma were taken and analysed for inflammatory mediators, with change in CSF IL-6 between 6 and 24 hours as the primary outcome measure. RESULTS: Six patients received IL-1Ra and seven received placebo. Concentrations of IL-6 in CSF and plasma were reduced by one standard deviation in the IL-1Ra group compared to the placebo group, between 6 and 24 hours, as predicted by the power calculation. This did not reach statistical significance (P = 0.08 and P = 0.06, respectively), since recruitment did not reach the target figure of 32. No adverse or serious adverse events reported were attributable to IL-1Ra. CONCLUSIONS: IL-1Ra appears safe in SAH patients. The concentration of IL-6 was lowered to the degree expected, in both CSF and plasma for patients treated with IL-1Ra.


Asunto(s)
Citocinas/líquido cefalorraquídeo , Proteína Antagonista del Receptor de Interleucina 1/administración & dosificación , Hemorragia Subaracnoidea/líquido cefalorraquídeo , Hemorragia Subaracnoidea/tratamiento farmacológico , Administración Intravenosa , Adulto , Anciano , Área Bajo la Curva , Citocinas/sangre , Método Doble Ciego , Ensayo de Inmunoadsorción Enzimática , Femenino , Escala de Coma de Glasgow , Humanos , Masculino , Persona de Mediana Edad , Hemorragia Subaracnoidea/sangre , Factores de Tiempo
16.
J Cereb Blood Flow Metab ; 34(3): 480-8, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24346689

RESUMEN

Acute-phase proteins (APPs) are key effectors of the immune response and are routinely used as biomarkers in cerebrovascular diseases, but their role during brain inflammation remains largely unknown. Elevated circulating levels of the acute-phase protein pentraxin-3 (PTX3) are associated with worse outcome in stroke patients. Here we show that PTX3 is expressed in neurons and glia in response to cerebral ischemia, and that the proinflammatory cytokine interleukin-1 (IL-1) is a key driver of PTX3 expression in the brain after experimental stroke. Gene deletion of PTX3 had no significant effects on acute ischemic brain injury. In contrast, the absence of PTX3 strongly compromised blood-brain barrier integrity and resolution of brain edema during recovery after ischemic injury. Compromised resolution of brain edema in PTX3-deficient mice was associated with impaired glial scar formation and alterations in scar-associated extracellular matrix production. Our results suggest that PTX3 expression induced by proinflammatory signals after ischemic brain injury is a critical effector of edema resolution and glial scar formation. This highlights the potential role for inflammatory molecules in brain recovery after injury and identifies APPs, in particular PTX3, as important targets in ischemic stroke and possibly other brain inflammatory disorders.


Asunto(s)
Edema Encefálico/metabolismo , Isquemia Encefálica/metabolismo , Proteína C-Reactiva/metabolismo , Cicatriz/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuroglía/metabolismo , Animales , Edema Encefálico/etiología , Edema Encefálico/inmunología , Edema Encefálico/patología , Isquemia Encefálica/complicaciones , Isquemia Encefálica/inmunología , Isquemia Encefálica/patología , Proteína C-Reactiva/genética , Proliferación Celular/efectos de los fármacos , Cicatriz/inmunología , Cicatriz/patología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Expresión Génica/efectos de los fármacos , Interleucina-1alfa/genética , Interleucina-1alfa/inmunología , Interleucina-1alfa/farmacología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-1beta/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Neuroglía/inmunología , Neuroglía/patología
17.
Dis Model Mech ; 6(4): 1043-8, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23519030

RESUMEN

Interleukin-1 (IL-1) is a key regulator of inflammation and ischaemic brain injury, but the contribution of central and peripheral sources of IL-1 to brain injury is not well understood. Here we show that haematopoietic-derived IL-1 is a key driver of ischaemic brain injury. Wild type (WT) mice transplanted with IL-1αß-deficient bone marrow displayed a significant (40%) reduction in brain injury induced by focal cerebral ischaemia compared with WT mice transplanted with WT bone marrow. This was paralleled by improved neurological outcome and the almost complete absence of splenic-derived, but not liver-derived, IL-1α after stroke in WT mice lacking haematopoietic-derived IL-1. IL-1αß knockout (KO) mice transplanted with IL-1αß-deficient bone marrow showed a 60% reduction in brain injury compared with WT mice receiving WT bone marrow. Transplantation of WT bone marrow in IL-1αß KO mice resulted in a similar level of blood-brain-barrier injury to that observed in WT mice receiving IL-1αß-deficient bone marrow. Cerebral oedema after brain injury was reduced in IL-1αß KO recipients irrespective of donor-derived IL-1, but a lack of haematopoetic IL-1 has also been associated with smaller brain oedema independently of recipient status. Thus, both central and haematopoietic-derived IL-1 are important contributors to brain injury after cerebral ischaemia. Identification of the cellular sources of IL-1 in the periphery could allow targeted interventions at these sites.


Asunto(s)
Lesiones Encefálicas/metabolismo , Isquemia Encefálica/metabolismo , Encéfalo/metabolismo , Sistema Hematopoyético/metabolismo , Interleucina-1alfa/metabolismo , Interleucina-1beta/metabolismo , Animales , Encéfalo/patología , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/patología , Isquemia Encefálica/complicaciones , Isquemia Encefálica/patología , Inflamación/complicaciones , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
18.
Glia ; 61(5): 813-24, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23404620

RESUMEN

Macrophage can adopt several phenotypes, process call polarization, which is crucial for shaping inflammatory responses to injury. It is not known if microglia, a resident brain macrophage population, polarizes in a similar way, and whether specific microglial phenotypes modulate cell death in response to brain injury. In this study, we show that both BV2-microglia and mouse bone marrow derived macrophages (BMDMs) were able to adopt different phenotypes after LPS (M1) or IL-4 (M2) treatment in vitro, but regulated cell death differently when added to mouse organotypic hippocampal brain slices. BMDMs induced cell death when added to control slices and exacerbated damage when combined with oxygen-glucose deprivation (OGD), independently of their phenotype. In contrast, vehicle- and M2-BV2-microglia were protective against OGD-induced death. Direct treatment of brain slices with IL-4 (without cell addition) was protective against OGD and induced an M2 phenotype in the slice. In vivo, intracerebral injection of LPS or IL-4 in mice induced microglial phenotypes similar to the phenotypes observed in brain slices and in cultured cells. After injury induced by middle cerebral artery occlusion, microglial cells did not adopt classical M1/M2 phenotypes, suggesting that another subtype of regulatory phenotype was induced. This study highlights functional differences between macrophages and microglia, in response to brain injury with fundamentally different outcomes, even if both populations were able to adopt M1 or M2 phenotypes. These data suggest that macrophages infiltrating the brain from the periphery after an injury may be cytotoxic, independently of their phenotype, while microglia may be protective.


Asunto(s)
Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Glucosa/deficiencia , Hipocampo/patología , Macrófagos/metabolismo , Macrófagos/patología , Microglía/metabolismo , Microglía/patología , Animales , Muerte Celular/fisiología , Hipoxia de la Célula/fisiología , Células Cultivadas , Hipocampo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Técnicas de Cultivo de Órganos , Especificidad de Órganos/fisiología
19.
PLoS One ; 8(2): e56441, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23418569

RESUMEN

PURPOSE: Neuroinflammation is involved in several brain disorders and can be monitored through expression of the translocator protein 18 kDa (TSPO) on activated microglia. In recent years, several new PET radioligands for TSPO have been evaluated in disease models. [(18)F]DPA-714 is a TSPO radiotracer with great promise; however results vary between different experimental models of neuroinflammation. To further examine the potential of [(18)F]DPA-714, it was compared directly to [(11)C]PK11195 in experimental cerebral ischaemia in rats. METHODS: Under anaesthesia, the middle cerebral artery of adult rats was occluded for 60 min using the filament model. Rats were allowed recovery for 5 to 7 days before one hour dynamic PET scans with [(11)C]PK11195 and/or [(18)F]DPA-714 under anaesthesia. RESULTS: Uptake of [(11)C]PK11195 vs [(18)F]DPA-714 in the ischemic lesion was similar (core/contralateral ratio: 2.84±0.67 vs 2.28±0.34 respectively), but severity of the brain ischemia and hence ligand uptake in the lesion appeared to vary greatly between animals scanned with [(11)C]PK11195 or with [(18)F]DPA-714. To solve this issue of inter-individual variability, we performed a direct comparison of [(11)C]PK11195 and [(18)F]DPA-714 by scanning the same animals sequentially with both tracers within 24 h. In this direct comparison, the core/contralateral ratio (3.35±1.21 vs 4.66±2.50 for [(11)C]PK11195 vs [(18)F]DPA-714 respectively) showed a significantly better signal-to-noise ratio (1.6 (1.3-1.9, 95%CI) fold by linear regression) for [(18)F]DPA-714. CONCLUSIONS: In a clinically relevant model of neuroinflammation, uptake for both radiotracers appeared to be similar at first, but a high variability was observed in our model. Therefore, to truly compare tracers in such models, we performed scans with both tracers in the same animals. By doing so, our result demonstrated that [(18)F]DPA-714 displayed a higher signal-to-noise ratio than [(11)C]PK11195. Our results suggest that, with the longer half-life of [(18)F] which facilitates distribution of the tracer across PET centre, [(18)F]DPA-714 is a good alternative for TSPO imaging.


Asunto(s)
Isquemia Encefálica/diagnóstico , Isoquinolinas , Tomografía de Emisión de Positrones/métodos , Pirazoles , Pirimidinas , Animales , Encéfalo/metabolismo , Encéfalo/patología , Isquemia Encefálica/diagnóstico por imagen , Antígeno CD11b/metabolismo , Radioisótopos de Carbono , Modelos Animales de Enfermedad , Radioisótopos de Flúor , Proteína Ácida Fibrilar de la Glía/metabolismo , Inmunohistoquímica , Masculino , Microglía/metabolismo , Microglía/patología , Proteínas Asociadas a Microtúbulos/metabolismo , Ratas , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Relación Señal-Ruido
20.
J Neurointerv Surg ; 5(6): 512-7, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22952245

RESUMEN

BACKGROUND: The mechanism of development of delayed cerebral ischemia (DCI) after aneurysmal subarachnoid hemorrhage (SAH) is poorly understood. Inflammatory processes are implicated in the development of ischemic stroke and may also predispose to the development of DCI following SAH. The objective of this study was to test whether concentrations of circulating inflammatory markers (C-reactive protein (CRP), interleukin-6 (IL-6) and interleukin 1 receptor antagonist (IL-1Ra)) were predictive for DCI following SAH. Secondary analyses considered white cell count (WCC) and erythrocyte sedimentation rate (ESR). METHODS: This was a single-center case-control study nested within a prospective cohort. Plasma inflammatory markers were measured in patients up to 15 days after SAH (initial, peak, average, final and rate of change to final). Cases were defined as those developing DCI. Inflammatory markers were compared between cases and randomly selected matched controls. RESULTS: Among the 179 participants there were 46 cases of DCI (26%). In primary analyses the rate of change of IL-6 was associated with DCI (OR 2.3 (95% CI 1.1 to 5.0); p=0.03). The final value and rate of change of WCC were associated with DCI (OR 1.2 (95% CI 1.0 to 1.3) and OR 1.3 (95% CI 1.0 to 1.6), respectively). High values of ESR were associated with DCI (OR 2.4 (95% CI 1.3 to 4.6) initial; OR 2.3 (95% CI 1.3 to 4.2) average; OR 2.1 (95% CI 1.1 to 3.9) peak; and OR 2.0 (95% CI 1.2 to 3.3) final value). CONCLUSIONS: Leucocytosis and change in IL-6 prior to DCI reflect impending cerebral ischemia. The time-independent association of ESR with DCI after SAH may identify this as a risk factor. These data suggest that systemic inflammatory mechanisms may increase the susceptibility to the development of DCI after SAH.


Asunto(s)
Isquemia Encefálica/etiología , Inflamación/patología , Hemorragia Subaracnoidea/complicaciones , Hemorragia Subaracnoidea/patología , Adulto , Anciano , Sedimentación Sanguínea , Isquemia Encefálica/patología , Proteína C-Reactiva/análisis , Estudios de Casos y Controles , Angiografía Cerebral , Estudios de Cohortes , Interpretación Estadística de Datos , Femenino , Humanos , Mediadores de Inflamación , Proteína Antagonista del Receptor de Interleucina 1/sangre , Interleucina-6/sangre , Recuento de Leucocitos , Leucocitosis/sangre , Leucocitosis/etiología , Masculino , Persona de Mediana Edad , Selección de Paciente , Valor Predictivo de las Pruebas , Pronóstico , Estudios Prospectivos , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA