Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 9 de 9
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Rev. esp. cardiol. (Ed. impr.) ; 75(9): 718-727, sept. 2022. tab
Article de Espagnol | IBECS | ID: ibc-208225

RÉSUMÉ

Introducción y objetivos La identificación de biomarcadores de fibrilación auricular (FA) subclínica en los pacientes con ictus criptogénico (ICr) es de gran interés. Con dicho objetivo, se evaluó el perfil de microARN circulante de los pacientes con ICr y FA frente a aquellos en ritmo sinusal. Métodos Se incluyó a 64 pacientes con ICr consecutivos monitorizados mediante Holter subcutáneo. Se seleccionó a 18 pacientes (9 con FA y 9 en ritmo sinusal persistente) para determinación de 754 microARN mediante tecnología de alto rendimiento. Se incluyó a 9 pacientes adicionales con ictus y FA concomitante para guiar la selección de microARN. Los microARN de interés se replicaron en una cohorte independiente (n=46). La asociación de biomarcadores con FA a los 6 y 12 meses se analizó mediante regresión logística. Resultados Ocho microARN mostraron expresión diferencial entre los pacientes con y sin FA. El miR-1-3p, un regulador génico involucrado en la arritmogénesis cardiaca, fue el único que permaneció significativamente más elevado en pacientes con ICr y FA de la cohorte de repetición, y además mostró una discreta asociación con la carga arrítmica. Los valores de miR-1-3p por encima de la mediana y la fracción de eyección de la aurícula izquierda se asociaron de forma independiente con la presencia de FA a los 6 y 12 meses. Conclusiones En nuestra cohorte, los valores plasmáticos de miR-1-3p fueron más altos en los pacientes con ICr y FA en el seguimiento. Nuestros resultados indican que el miR-1-3p podría ser un nuevo biomarcador de FA oculta en los pacientes con ICr (AU)


Introduction and objectives Identifying biomarkers of subclinical atrial fibrillation (AF) is of most interest in patients with cryptogenic stroke (CrS). We sought to evaluate the circulating microRNA (miRNA) profile of patients with CrS and AF compared with those in persistent sinus rhythm. Methods Among 64 consecutive patients with CrS under continuous monitoring by a predischarge insertable monitor, 18 patients (9 with AF and 9 in persistent sinus rhythm) were selected for high-throughput determination of 754 miRNAs. Nine patients with concomitant stroke and AF were also screened to improve the yield of miRNA selection. Differentially expressed miRNAs were replicated in an independent cohort (n=46). Biological markers were stratified by the median and included in logistic regression analyses to evaluate their association with AF at 6 and 12 months. Results Eight miRNAs were differentially expressed between patients with and without AF. In the replication cohort, miR-1-3p, a gene regulator involved in cardiac arrhythmogenesis, was the only miRNA to remain significantly higher in patients with CrS and AF vs those in sinus rhythm and showed a modest association with AF burden. High (= above the median) miR-1-3p plasma values, together with a low left atrial ejection fraction, were independently associated with the presence of AF at 6 and 12 months. Conclusions In this cohort, plasma levels of miR-1-3p were elevated in CrS patients with subsequent AF. Our results preliminarily suggest that miR-1-3p could be a novel biomarker that, together with clinical parameters, could help identify patients with CrS and a high risk of occult AF (AU)


Sujet(s)
Humains , Mâle , Femelle , Adulte d'âge moyen , Sujet âgé , Fibrillation auriculaire , microARN/génétique , Études cas-témoins , Études prospectives , Fibrillation auriculaire/complications , Fibrillation auriculaire/diagnostic , Fibrillation auriculaire/génétique , Marqueurs biologiques
2.
Cell Biosci ; 11(1): 89, 2021 May 17.
Article de Anglais | MEDLINE | ID: mdl-34001233

RÉSUMÉ

BACKGROUND: Iron deficiency (ID) is common in patients with heart failure (HF) and is associated with poor outcomes, yet its role in the pathophysiology of HF is not well-defined. We sought to determine the consequences of HF neurohormonal activation in iron homeostasis and mitochondrial function in cardiac cells. METHODS: HF was induced in C57BL/6 mice by using isoproterenol osmotic pumps and embryonic rat heart-derived H9c2 cells were subsequently challenged with Angiotensin II and/or Norepinephrine. The expression of several genes and proteins related to intracellular iron metabolism were assessed by Real time-PCR and immunoblotting, respectively. The intracellular iron levels were also determined. Mitochondrial function was analyzed by studying the mitochondrial membrane potential, the accumulation of radical oxygen species (ROS) and the adenosine triphosphate (ATP) production. RESULTS: Hearts from isoproterenol-stimulated mice showed a decreased in both mRNA and protein levels of iron regulatory proteins, transferrin receptor 1, ferroportin 1 and hepcidin compared to control mice. Furthermore, mitoferrin 2 and mitochondrial ferritin were also downregulated in the hearts from HF mice. Similar data regarding these key iron regulatory molecules were found in the H9c2 cells challenged with neurohormonal stimuli. Accordingly, a depletion of intracellular iron levels was found in the stimulated cells compared to non-stimulated cells, as well as in the hearts from the isoproterenol-induced HF mice. Finally, neurohormonal activation impaired mitochondrial function as indicated by the accumulation of ROS, the impaired mitochondrial membrane potential and the decrease in the ATP levels in the cardiac cells. CONCLUSIONS: HF characteristic neurohormonal activation induced changes in the regulation of key molecules involved in iron homeostasis, reduced intracellular iron levels and impaired mitochondrial function. The current results suggest that iron could be involved in the pathophysiology of HF.

3.
Histol Histopathol ; 28(1): 43-51, 2013 01.
Article de Anglais | MEDLINE | ID: mdl-23233058

RÉSUMÉ

Alzheimer's disease (AD), the most prevalent neurodegenerative disease worldwide, has two main hallmarks: extracellular deposits of amyloid ß-peptide (Aß) and intracellular neurofibrillary tangles composed by tau protein. Most AD cases are sporadic and are not dependent on known genetic causes; aging is the major risk factor for AD. Therefore, the oxidative stress has been proposed to initiate the uncontrolled increase in Aß production and also to mediate the Aß's deleterious effects on brain cells, especially on neurons from the cortex and hippocampus. The production of free radicals in the presence of nitric oxide (NO) yields to the peroxynitrite generation, a very reactive agent that nitrotyrosinates the proteins irreversibly. The nitrotyrosination produces a loss of protein physiological functions, contributing to accelerate AD progression. One of the most nitrotyrosinated proteins in AD is the enzyme triosephosphate isomerase (TPI) that isomerises trioses, regulating glucose consumption by both phosphate pentose and glycolytic pathways and thereby pyruvate production. Hence, any disturbance in the glucose supply could affect the proper brain function, considering that the brain has a high rate of glucose consumption. Besides this directly affecting to the energetic metabolism of the neurons, TPI modifications, such as mutation or nitrotyrosination, increase methylglyoxal production, a toxic precursor of advanced glycated end-products (AGEs) and responsible for protein glycation. Moreover, nitro-TPI aggregates interact with tau protein inducing the intraneuronal aggregation of tau. Here we review the relationship between modified TPI and AD, highlighting the relevance of this protein in AD pathology and the consequences of protein nitro-oxidative modifications.


Sujet(s)
Maladie d'Alzheimer/enzymologie , Maladie d'Alzheimer/physiopathologie , Triose phosphate isomerase/métabolisme , Humains , Nitrosation/physiologie , Stress oxydatif/physiologie
4.
Rev Neurol ; 52(10): 618-22, 2011 May 16.
Article de Espagnol | MEDLINE | ID: mdl-21488009

RÉSUMÉ

Senescence-accelerated mice (SAM) represent an aging model establish by selective inbreeding of the AKR/J strain. SAMP8 is a suitable model to study the genetics or proteics fundamental mechanisms of aging, in physiological or pathological conditions, because SAMP8 develop neuropathological markers also found in neurodegenerative diseases like Alzheimer. Melatonin is known as sleep hormone because its action controlling the sleep/awake circadian rhythm. Moreover, melatonin has antioxidant properties and may have an important anti-aging role. The chronic treatment with melatonin in the SAMP8 model was able to reduce oxidative stress and the neurodegenerative calpain/Cdk5 pathway and primed phosphorylation of GSK3beta and tau hiperphosphorylation markers of cerebral aging and neurodegeneration in SAMP8 brains, indicating the neuroprotective and anti-aging effect of melatonin.


Sujet(s)
Vieillissement/effets des médicaments et des substances chimiques , Mélatonine/pharmacologie , Mélatonine/usage thérapeutique , Lignées consanguines de souris , Maladies neurodégénératives/traitement médicamenteux , Vieillissement/anatomopathologie , Vieillissement/physiologie , Animaux , Antioxydants/pharmacologie , Antioxydants/usage thérapeutique , Marqueurs biologiques/métabolisme , Modèles animaux de maladie humaine , Glycogen Synthase Kinase 3/métabolisme , Glycogen synthase kinase 3 beta , Souris , Maladies neurodégénératives/anatomopathologie , Maladies neurodégénératives/physiopathologie , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/usage thérapeutique , Stress oxydatif/effets des médicaments et des substances chimiques , Protéines tau/métabolisme
5.
Exp Gerontol ; 45(9): 702-10, 2010 Sep.
Article de Anglais | MEDLINE | ID: mdl-20460146

RÉSUMÉ

Dietary interventions have been proposed as a way to increase lifespan and improve health. The senescence-accelerated prone 8 (SAMP8) mice have a shorter lifespan and show alterations in the central nervous system. Moreover, this mouse strain shows decreased sirtuin 1 protein expression and elevated expression of the acetylated targets NFkappaB and FoxO1, which are implicated in transcriptional control of key genes in cell proliferation and cell survival, in reference to control strain, SAMR1. After eight weeks of intermittent fasting, sirtuin 1 protein expression was recovered in SAMP8. This recovery was accompanied by a reduction in the two acetylated targets. Furthermore, SAMP8 showed a lower protein expression of BDNF and HSP70 while intermittent fasting re-established normal values. The activation of JNK and FoxO1 was also reduced in SAMP8 mice subjected to an IF regimen, compared with control SAMP8. Our findings provide new insights into the participation of sirtuin 1 in ageing and point to a potential novel application of this enzyme to prevent frailty due to ageing processes in the brain.


Sujet(s)
Vieillissement/physiologie , Jeûne/physiologie , Lignées consanguines de souris/génétique , Neuroprotecteurs , Animaux , Poids , Encéphale/croissance et développement , Encéphale/physiologie , Ration calorique , Colorants fluorescents , Régulation de l'expression des gènes au cours du développement , Mâle , Souris , Lignées consanguines de souris/physiologie , Séquençage par oligonucléotides en batterie , ARN/génétique , ARN/isolement et purification , RT-PCR , Sirtuine-1/physiologie , Transcription génétique
6.
J Pineal Res ; 47(3): 228-37, 2009 Oct.
Article de Anglais | MEDLINE | ID: mdl-19650880

RÉSUMÉ

Sirtuin 1 is a member of the sirtuin family of protein deacetylases, which have attracted considerable attention as mediators of lifespan extension in several model organisms. Induction of sirtuin 1 expression also attenuates neuronal degeneration and death in animal models of Alzheimer's disease and Huntington's disease. In this study, an in vitro model of neuronal aging was used to test in several ways whether melatonin acts as a sirtuin 1 inducer and if this effect could be neuroprotective. It is shown that melatonin is able to increase the level of this deacetylase in young primary neurons, as well as in aged neurons. We also observed an increase in the deacetylation of several substrates of sirtuin 1, such as p53, PGC-1alpha, FoxO1, ADAM10 and NFkappaB. In addition, there was a reduction in its nuclear translocation and, subsequently, an improvement in transcriptional activity. Sirtinol, a sirtuin 1 inhibitor, was used to correlate these effects with sirtuin. It is shown that sirtinol reduces sirtuin 1 expression and impairs the beneficial action of melatonin on cell viability and apoptosis prevention. Moreover, some of the sirtuin 1 substrates studied also reversed the melatonin effect when sirtinol is added to the cells, mainly p53. Globally, these results add weight to the findings of previous reports, indicating a new role for melatonin in improving cell function gated to an increased neuroprotective role for the sirtuin 1 pathway.


Sujet(s)
Antioxydants/pharmacologie , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Mélatonine/pharmacologie , Sirtuine-1/métabolisme , Protéines ADAM/métabolisme , Protéine ADAM10 , Animaux , Technique de Western , Cellules cultivées , Facteurs de transcription Forkhead/métabolisme , Immunoprécipitation , Facteur de transcription NF-kappa B/métabolisme , Protéines de tissu nerveux/métabolisme , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes , Protéines de liaison à l'ARN/métabolisme , Rats , Rat Sprague-Dawley , RT-PCR , Transduction du signal/effets des médicaments et des substances chimiques , Facteurs de transcription/métabolisme , Protéine p53 suppresseur de tumeur/métabolisme
7.
Neuroscience ; 154(4): 1388-97, 2008 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-18538940

RÉSUMÉ

We examined the expression of SIRT1 in several experimental paradigms of human pathologies. We used a neuroblastoma cell line (B65), neuronal primary cultures (hippocampus and cerebellar granule cells) and in vivo approaches in rat and senescence murine models (SAM). Cell cultures and rats were treated with several well-know neurotoxins, i.e. rotenone, MPP(+), kainate and 3-nitropropionic acid. Subsequently, SIRT1 expression was compared in these different paradigms of neurotoxicity. The pattern of expression of SIRT1 in proliferating cell cultures (B65) was different to that in quiescent cell cultures. In the murine model of senescence (senescence-accelerated mice prone, SAMP8), SIRT1 expression progressively decreased, while in the control strain (senescence-accelerated mice resistant, SAMR1) it increased. Finally, we studied human samples of Parkinson's disease (PD), dementia with Lewy bodies (DLB) and Huntington's diseases (HD). SIRT1 expression decreased dramatically in HD, but there were no significant changes in Parkinson-related illnesses. In conclusion, SIRT1 expression may be a good sensor of toxic neuronal processes.


Sujet(s)
Vieillissement/métabolisme , Maladies neurodégénératives/métabolisme , Sirtuines/métabolisme , Animaux , Apoptose/effets des médicaments et des substances chimiques , Apoptose/physiologie , Technique de Western , Cellules cultivées , Cytométrie en flux , Humains , Mâle , Souris , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme , Neurotoxines/pharmacologie , Rats , Rat Sprague-Dawley , Sirtuine-1 , Sirtuines/effets des médicaments et des substances chimiques
8.
Neuroscience ; 146(1): 350-65, 2007 Apr 25.
Article de Anglais | MEDLINE | ID: mdl-17343987

RÉSUMÉ

The biochemical pathways involved in neuronal cell death in Parkinson's disease are not completely characterized. Mitochondrial dysfunction, specifically alteration of the mitochondrial complex I, is the primary target of the parkinsonian neurotoxin 1-methyl-4-phenylpyridinium (MPP+) induced apoptosis in neurons. In the present study, we examine the role of caspase-dependent and -independent routes in MPP+-induced apoptosis in rat cerebellar granule neurons (CGNs). We show a distinct increase in the expression of the cell cycle proteins cyclin D, cyclin E, cdk2, cdk4 and the transcription factor E2F-1 following a MPP+ treatment of CGNs. Flavopiridol (FLAV), a broad inhibitor of cyclin-dependent kinases (CDKs), attenuated the neurotoxic effects of MPP+ and significantly attenuates apoptosis mediated by MPP+ 200 microM. Likewise, the antioxidant vitamin E (vit E) increases neuronal cell viability and attenuates apoptosis induced by MPP+. Moreover, the expression levels of cyclin D and E2F-1 induced by this parkinsonian neurotoxin were also attenuated by vit E. Since, the broad-spectrum caspase inhibitor zVAD-fmk did not attenuate MPP+-induced apoptosis in CGNs, our data provide a caspase-independent mechanism mediated by neuronal reentry in the cell cycle and increased expression of the pro-apoptotic transcription factor E2F-1. Our results also suggest a potential role of oxidative stress in neuronal reentry in the cell cycle mediated by MPP+. Finally, our data further support the therapeutic potential of flavopiridol, for the treatment of Parkinson's disease.


Sujet(s)
1-Méthyl-4-phényl-pyridinium/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Kinases cyclines-dépendantes/métabolisme , Herbicides/pharmacologie , Neurones/effets des médicaments et des substances chimiques , Analyse de variance , Animaux , Animaux nouveau-nés , Caspases/métabolisme , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Cervelet/cytologie , Relation dose-effet des médicaments , Facteur de transcription E2F1/métabolisme , Antienzymes/pharmacologie , Cytométrie en flux , Rats , Espèces réactives de l'oxygène/métabolisme , Roténone/pharmacologie , Facteurs temps
9.
J Neural Transm (Vienna) ; 114(4): 405-16, 2007.
Article de Anglais | MEDLINE | ID: mdl-16906355

RÉSUMÉ

Li(+) exerts protective effect against several neurotoxins in neuronal cell preparations. Here we examined the antiapoptotic effects of GSK3beta in cerebellar granule neurons (CGNs) in the presence of several neurotoxins. Acute treatment with Li(+) protected neurons against nocodazole and serum/potassium (S/K) deprivation, but were ineffective against kainic acid and MPP(+). Li(+) 5 mM also decreased caspase-3 activation induced by nocodazole and S/K deprivation as measured by Ac-DEVD-p-nitroaniline and the breakdown of alpha-spectrin. All the neurotoxins used in the present study activated GSK3beta, evaluated with a specific antibody phospho-GSK-3beta (Ser9) by Western-blot and immunocytochemistry and were always inhibited by Li(+) 5 mM. Our results implicate Li(+) in the regulation of apoptosis mediated by caspase activation (Type I). Furthermore inhibition of GSK3beta by acute treatment with Li(+) 5 mM is not an indicator of neuroprotection. The acute antiapoptotic function of Li(+) is discussed in terms of its inhibition of Type I pathway, the intrinsic (mitochondrial) apoptotic pathway in cerebellar granule cells.


Sujet(s)
Apoptose/effets des médicaments et des substances chimiques , Chlorure de lithium/pharmacologie , Neurones/effets des médicaments et des substances chimiques , Neuroprotecteurs/pharmacologie , 1-Méthyl-4-phényl-1,2,3,6-tétrahydropyridine/pharmacologie , Animaux , Caspases/effets des médicaments et des substances chimiques , Caspases/métabolisme , Cellules cultivées , Agents dopaminergiques/toxicité , Activation enzymatique/effets des médicaments et des substances chimiques , Agonistes des acides aminés excitateurs/toxicité , Cytométrie en flux , Glycogen Synthase Kinase 3/métabolisme , Glycogen synthase kinase 3 beta , Immunohistochimie , Acide kaïnique/toxicité , Neurones/métabolisme , Neurones/anatomopathologie , Nocodazole/toxicité , Rats , Rat Sprague-Dawley
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE