Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Sci Total Environ ; 804: 150117, 2022 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-34508938

RESUMEN

Plastic debris into the environment is a growing threat for the ecosystems and human health. The seafood sector is particularly concerned because it generates plastic losses and can be endangered by plastic contamination. Life cycle assessment (LCA) does not properly consider plastic losses and related impacts, which is a problem in order to find relevant mitigation strategies without burden shifting. This work proposes a methodology for quantifying flows of plastics from the life cycle of the seafood products to the environment. It is based on loss rate and final release rate considering a pre-fate approach as proposed by the Plastic Leak Project. They are defined for 5 types of micro and macro plastic losses: lost fishing gears, marine coatings, plastic pellets, tire abrasion and plastic mismanaged at the end-of-life. The methodology is validated with a case study applied to French fish products for which relevant data are available in the Agribalyse 3.0 database. Results show that average plastic losses are from 75 mg to 4345 mg per kg of fish at the consumer, depending on the species and the related fishing method. The main plastic losses come from lost fishing gears (macroplastics) and tire abrasion (microplastics). Results show high variability: when mismanaged, plastic packaging at the end-of-life (macroplastics) is the main loss to the environment. As a next step the methodology is to be applied to other fish or shellfish products, or directly implemented in a life cycle inventory database. Further research should characterize the related impacts to the environment when life cycle impact assessment methodologies will be available, and identify eco-design solutions to decrease the major flows to the environment identified.


Asunto(s)
Plásticos , Contaminantes Químicos del Agua , Animales , Ecosistema , Monitoreo del Ambiente , Productos Pesqueros , Humanos , Estadios del Ciclo de Vida , Alimentos Marinos , Residuos/análisis , Contaminantes Químicos del Agua/análisis
2.
Acta Neuropathol ; 137(4): 599-617, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30721409

RESUMEN

Brains of Alzheimer's disease patients are characterized by the presence of amyloid plaques and neurofibrillary tangles, both invariably associated with neuroinflammation. A crucial role for NLRP3-ASC inflammasome [NACHT, LRR and PYD domains-containing protein 3 (NLRP3)-Apoptosis-associated speck-like protein containing a CARD (ASC)] in amyloid-beta (Aß)-induced microgliosis and Aß pathology has been unequivocally identified. Aß aggregates activate NLRP3-ASC inflammasome (Halle et al. in Nat Immunol 9:857-865, 2008) and conversely NLRP3-ASC inflammasome activation exacerbates amyloid pathology in vivo (Heneka et al. in Nature 493:674-678, 2013), including by prion-like ASC-speck cross-seeding (Venegas et al. in Nature 552:355-361, 2017). However, the link between inflammasome activation, as crucial sensor of innate immunity, and Tau remains unexplored. Here, we analyzed whether Tau aggregates acting as prion-like Tau seeds can activate NLRP3-ASC inflammasome. We demonstrate that Tau seeds activate NLRP3-ASC-dependent inflammasome in primary microglia, following microglial uptake and lysosomal sorting of Tau seeds. Next, we analyzed the role of inflammasome activation in prion-like or templated seeding of Tau pathology and found significant inhibition of exogenously seeded Tau pathology by ASC deficiency in Tau transgenic mice. We furthermore demonstrate that chronic intracerebral administration of the NLRP3 inhibitor, MCC950, inhibits exogenously seeded Tau pathology. Finally, ASC deficiency also decreased non-exogenously seeded Tau pathology in Tau transgenic mice. Overall our findings demonstrate that Tau-seeding competent, aggregated Tau activates the ASC inflammasome through the NLRP3-ASC axis, and we demonstrate an exacerbating role of the NLRP3-ASC axis on exogenously and non-exogenously seeded Tau pathology in Tau mice in vivo. The NLRP3-ASC inflammasome, which is an important sensor of innate immunity and intensively explored for its role in health and disease, hence presents as an interesting therapeutic approach to target three crucial pathogenetic processes in AD, including prion-like seeding of Tau pathology, Aß pathology and neuroinflammation.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Proteínas Adaptadoras de Señalización CARD/metabolismo , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Agregado de Proteínas/fisiología , Proteínas tau/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Gliosis/genética , Gliosis/metabolismo , Gliosis/patología , Interleucina-1beta/metabolismo , Ratones , Ratones Transgénicos , Microglía/metabolismo , Microglía/patología , Proteínas tau/genética
3.
J Neuroinflammation ; 13: 20, 2016 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-26818951

RESUMEN

BACKGROUND: The proinflammatory cytokine interleukin-1ß (IL-1ß) is overexpressed in Alzheimer disease (AD) as a key regulator of neuroinflammation. Amyloid-ß (Aß) peptide triggers activation of inflammasomes, protein complexes responsible for IL-1ß maturation in microglial cells. Downregulation of NALP3 (NACHT, LRR, and PYD domains-containing protein 3) inflammasome has been shown to decrease amyloid load and rescue cognitive deficits in a mouse model of AD. Whereas activation of inflammasome in microglial cells has been described in AD, no data are currently available concerning activation of inflammasome in astrocytes, although they are involved in inflammatory response and phagocytosis. Here, by targeting the inflammasome adaptor protein ASC (apoptosis-associated speck-like protein containing a CARD domain), we investigated the influence of activation of the inflammasome on the phagocytic activity of astrocytes. METHODS: We used an ASC knockout mouse model, as ASC is a central protein in the inflammasome, acting as an adaptor and stabilizer of the complex and thus critical for its activation. Lipopolysaccharide (LPS)-primed primary cultures of astrocytes from newborn mice were utilized to evaluate Aß-induced inflammasome activation by measuring IL-1ß release by ECLIA (electro-chemiluminescence immunoassay). Phagocytosis efficiency was measured by incorporation of bioparticles, and the release of the chemokine CCL3 (C-C motif ligand 3) was measured by ECLIA. ASC mice were crossbred with 5xFAD (familial Alzheimer disease) mice and tested for spatial reference memory using the Morris water maze (MWM) at 7-8 months of age. Amyloid load and CCL3 were quantified by thioflavine S staining and quantitative real-time polymerase chain reaction (qRT-PCR), respectively. RESULTS: Cultured astrocytes primed with LPS and treated with Aß showed an ASC-dependent production of IL-1ß resulting from inflammasome activation mediated by Aß phagocytosis and cathepsin B enzymatic activity. ASC+/- astrocytes displayed a higher phagocytic activity as compared to ASC+/+ and ASC -/- cells, resulting from a higher release of the chemokine CCL3. A significant decrease in amyloid load was measured in the brain of 7-8-month-old 5xFAD mice carrying the ASC +/- genotype, correlated with an increase in CCL3 gene expression. In addition, the ASC +/- genotype rescued spatial reference memory deficits observed in 5xFAD mice. CONCLUSIONS: Our results demonstrate that Aß is able to activate astrocytic inflammasome. Downregulation of inflammasome activity increases phagocytosis in astrocytes due to the release of CCL3. This could explain why downregulation of inflammasome activity decreases amyloid load and rescues memory deficits in a mouse model of AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Proteínas Reguladoras de la Apoptosis/metabolismo , Astrocitos/metabolismo , Fagocitos/metabolismo , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/farmacología , Precursor de Proteína beta-Amiloide/genética , Animales , Animales Recién Nacidos , Proteínas Reguladoras de la Apoptosis/genética , Astrocitos/efectos de los fármacos , Proteínas Adaptadoras de Señalización CARD , Estudios de Casos y Controles , Células Cultivadas , Quimiocina CCL3/metabolismo , Quimiocina CCL3/farmacología , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Humanos , Ionóforos/farmacología , Trastornos de la Memoria/etiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Nigericina/farmacología , Fragmentos de Péptidos/farmacología , Presenilina-1/genética
4.
FASEB J ; 28(6): 2620-31, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24604080

RESUMEN

Tau alterations are now considered an executor of neuronal demise and cognitive dysfunction in Alzheimer's disease (AD). Mouse models combining amyloidosis and tauopathy and their parental counterparts are important tools to further investigate the interplay of abnormal amyloid-ß (Aß) and Tau species in pathogenesis, synaptic and neuronal dysfunction, and cognitive decline. Here, we crossed APP/PS1 mice with 5 early-onset familial AD mutations (5xFAD) and TauP301S (PS19) transgenic mice, denoted F(+)/T(+) mice, and phenotypically compared them to their respective parental strains, denoted F(+)/T(-) and F(-)/T(+) respectively, as controls. We found dramatically aggravated tauopathy (~10-fold) in F(+)/T(+) mice compared to the parental F(-)/T(+) mice. In contrast, amyloidosis was unaltered compared to the parental F(+)/T(-) mice. Tauopathy was invariably and very robustly aggravated in hippocampal and cortical brain regions. Most important, F(+)/T(+) displayed aggravated cognitive deficits in a hippocampus-dependent spatial navigation task, compared to the parental F(+)/T(-) strain, while parental F(-)/T(+) mice did not display cognitive impairment. Basal synaptic transmission was impaired in F(+)/T(+) mice compared to nontransgenic mice and the parental strains (≥40%). Finally, F(+)/T(+) mice displayed a significant hippocampal atrophy (~20%) compared to nontransgenic mice, in contrast to the parental strains. Our data indicate for the first time that pathological Aß species (or APP/PS1) induced changes in Tau contribute to cognitive deficits correlating with synaptic deficits and hippocampal atrophy in an AD model. Our data lend support to the amyloid cascade hypothesis with a role of pathological Aß species as initiator and pathological Tau species as executor.


Asunto(s)
Enfermedad de Alzheimer/patología , Trastornos del Conocimiento/etiología , Transmisión Sináptica , Tauopatías/complicaciones , Péptidos beta-Amiloides , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Atrofia/patología , Trastornos del Conocimiento/patología , Modelos Animales de Enfermedad , Femenino , Glucógeno Sintasa Quinasa 3/metabolismo , Hipocampo/patología , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Transgénicos , Presenilina-1/genética , Tauopatías/patología , Proteínas tau/genética
5.
Curr Alzheimer Res ; 9(3): 344-60, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22272616

RESUMEN

The double-stranded RNA-dependent protein kinase (PKR) is switched on by a wide range of stimuli, including the amyloid peptide. Then, PKR transmits signals to the translational machinery, apoptosis and inflammatory signaling pathways by interacting with some adapters. In virus-infected cells, PKR engages the nucleus factor κB (NF-κB) pathway. In many models of Alzheimer's disease (AD) and patients with AD, PKR was activated. Furthermore, there is strong evidence implicating the inflammatory process in the AD brain. However, the PKR involvement in inflammatory responses in AD is not elucidated. Based on our previous in vitro results, the aim of this study was to evaluate the effects of a pharmacological inhibition of PKR in inflammation in APPswePS1dE9 transgenic mice. Our results showed that PKR inhibition prevented the NF-κB activation and production of tumor necrosis factor alpha (TNFα) and interleukin (IL)-1ß at 12 months of age without decrease of Aß42 levels and memory deficits. Surprisingly, PKR inhibition failed to prevent IL-1ß- mediated inflammation and induced a great increase in ß-amyloid peptide (Aß42) levels at 18 months of age. In this model, our findings highlight the lack of relationship between inflammation and Aß42 levels. Moreover, the age-dependent inflammatory response must be carefully taken into account in the establishment of an anti-inflammatory therapy in AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Fragmentos de Péptidos/metabolismo , Regulación hacia Arriba/genética , eIF-2 Quinasa/antagonistas & inhibidores , Envejecimiento/genética , Envejecimiento/patología , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/genética , Animales , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Encéfalo/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Transgénicos , Fragmentos de Péptidos/genética , eIF-2 Quinasa/metabolismo
6.
J Biol Chem ; 285(2): 1272-82, 2010 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-19889624

RESUMEN

For 10 years, research has focused on signaling pathways controlling translation to explain neuronal death in Alzheimer Disease (AD). Previous studies demonstrated in different cellular and animal models and AD patients that translation is down-regulated by the activation of double-stranded RNA-dependent protein kinase (PKR). Among downstream factors of PKR, the Fas-associated protein with a death domain (FADD) and subsequent activated caspase-8 are responsible for PKR-induced apoptosis in recombinant virus-infected cells. However, no studies have reported the role of PKR in death receptor signaling in AD. The aim of this project is to determine physical and functional interactions of PKR with FADD in amyloid-beta peptide (Abeta) neurotoxicity and in APP(SL)PS1 KI transgenic mice. In SH-SY5Y cells, results showed that Abeta42 induced a large increase in phosphorylated PKR and FADD levels and a physical interaction between PKR and FADD in the nucleus, also observed in the cortex of APP(SL)PS1 KI mice. However, PKR gene silencing or treatment with a specific PKR inhibitor significantly prevented the increase in pT(451)-PKR and pS(194)-FADD levels in SH-SY5Y nuclei and completely inhibited activities of caspase-3 and -8. The contribution of PKR in neurodegeneration through the death receptor signaling pathway may support the development of therapeutics targeting PKR to limit neuronal death in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/farmacología , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Transducción de Señal/efectos de los fármacos , eIF-2 Quinasa/metabolismo , Enfermedad de Alzheimer/inducido químicamente , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Animales , Caspasa 3/genética , Caspasa 3/metabolismo , Caspasa 8/genética , Caspasa 8/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Corteza Cerebral/metabolismo , Inhibidores Enzimáticos/farmacología , Proteína de Dominio de Muerte Asociada a Fas/genética , Ratones , Ratones Transgénicos , Fosforilación/efectos de los fármacos , Fosforilación/genética , Transducción de Señal/genética , eIF-2 Quinasa/antagonistas & inhibidores , eIF-2 Quinasa/genética
7.
J Alzheimers Dis ; 21(4): 1217-31, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21504114

RESUMEN

Alzheimer's disease (AD), a neurodegenerative disorder, is the most common form of dementia in the elderly individuals. Among the pathogenic mechanisms in AD, chronic systemic inflammation is described and characterized by massive production of proinflammatory cytokines by peripheral blood mononuclear cells (PBMCs), which may contribute to an altered immune response and exacerbation of neurodegeneration. Studies have also reported increased double-stranded RNA-dependent protein kinase (PKR) activation in the PBMCs of patients with AD. Interestingly, PKR could be involved in NF-κB activation, leading to production of a wide range of cytokines. We proposed to decrease proinflammatory cytokines production and release by treating the PBMCs in 25 patients with AD with a specific inhibitor of PKR. Our results showed that PKR inhibition greatly decreased tumor necrosis factor , interleukin (IL)-1α, IL-1ß, and IL-6 production and release but did not affect the chemokine RANTES. Moreover, inhibition of the proinflammatory factors was correlated with prevention of caspase-3 activation. These results indicated that specific inhibition of PKR at the peripheral level might decrease the inflammatory response in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Citocinas/antagonistas & inhibidores , Leucocitos Mononucleares/enzimología , Inhibidores de Proteínas Quinasas/farmacología , eIF-2 Quinasa/antagonistas & inhibidores , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/patología , Caspasa 3/metabolismo , Inhibidores de Caspasas , Línea Celular Tumoral , Supervivencia Celular/fisiología , Células Cultivadas , Quimiocina CCL5/biosíntesis , Quimiocina CCL5/metabolismo , Citocinas/biosíntesis , Citocinas/metabolismo , Femenino , Humanos , Inflamación/enzimología , Inflamación/patología , Inflamación/prevención & control , Leucocitos Mononucleares/efectos de los fármacos , Masculino , FN-kappa B/antagonistas & inhibidores , FN-kappa B/fisiología , eIF-2 Quinasa/metabolismo
8.
J Cell Mol Med ; 13(8A): 1476-88, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19602051

RESUMEN

Amyloid beta-peptide (Abeta) deposits and neurofibrillary tangles are key hallmarks in Alzheimer's disease (AD). Abeta stimulates many signal transducers involved in the neuronal death. However, many mechanisms remain to be elucidated because no definitive therapy of AD exists. Some studies have focused on the control of translation which involves eIF2 and eIF4E, main eukaryotic factors of initiation. The availability of these factors depends on the activation of the double-stranded RNA-dependent protein kinase (PKR) and the mammalian target of rapamycin (mTOR), respectively. mTOR positively regulates the translation while PKR results in a protein synthesis shutdown. Many studies demonstrated that the PKR signalling pathway is up-regulated in cellular and animal models of AD and in the brain of AD patients. Interestingly, our results showed that phosphorylated PKR and eIF2alpha levels were significantly increased in lymphocytes of AD patients. These modifications were significantly correlated with cognitive and memory test scores performed in AD patients. On the contrary, the mTOR signalling pathway is down-regulated in cellular and animal models of AD. Recently, we showed that p53, regulated protein in development and DNA damage response 1 and tuberous sclerosis complex 2 could represent molecular links between PKR and mTOR signalling pathways. PKR could be an early biomarker of the neuronal death and a critical target for a therapeutic programme in AD.


Asunto(s)
Enfermedad de Alzheimer/enzimología , eIF-2 Quinasa/metabolismo , Enfermedad de Alzheimer/diagnóstico , Animales , Biomarcadores/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Biosíntesis de Proteínas , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR
9.
Neurobiol Dis ; 36(1): 151-61, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19631745

RESUMEN

The control of translation is disturbed in Alzheimer's disease (AD). This study analysed the crosslink between the up regulation of double-stranded RNA-dependent-protein kinase (PKR) and the down regulation of mammalian target of rapamycin (mTOR) signalling pathways via p53, the protein Regulated in the Development and DNA damage response 1 (Redd1) and the tuberous sclerosis complex (TSC2) factors in two beta-amyloid peptide (Abeta) neurotoxicity models. In SH-SY5Y cells, Abeta42 induced an increase of P(T451)-PKR and of the ratio p66/(p66+p53) in nuclei and a physical interaction between these proteins. Redd1 gene levels increased and P(T1462)-TSC2 decreased. These disturbances were earlier in rat primary neurons with nuclear co-localization of Redd1 and PKR. The PKR gene silencing in SH-SY5Y cells prevented these alterations. p53, Redd1 and TSC2 could represent the molecular links between PKR and mTOR in Abeta neurotoxicity. PKR could be a critical target in a therapeutic program of AD.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Neuronas/efectos de los fármacos , Fragmentos de Péptidos/toxicidad , Proteínas Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Supresoras de Tumor/metabolismo , eIF-2 Quinasa/metabolismo , Análisis de Varianza , Animales , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Células Cultivadas , Corteza Cerebral/citología , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Embrión de Mamíferos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Humanos , Inmunoprecipitación/métodos , Neuroblastoma/patología , Neuronas/citología , Fosforilación/efectos de los fármacos , Proteínas Quinasas/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Wistar , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Serina-Treonina Quinasas TOR , Factores de Transcripción , Transfección/métodos , Proteína 2 del Complejo de la Esclerosis Tuberosa , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/genética , eIF-2 Quinasa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA