Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 23(9)2022 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-35563362

RESUMEN

The pathological accumulation of parenchymal and vascular amyloid-beta (Aß) are the main hallmarks of Alzheimer's disease (AD) and Cerebral Amyloid Angiopathy (CAA), respectively. Emerging evidence raises an important contribution of vascular dysfunction in AD pathology that could partially explain the failure of anti-Aß therapies in this field. Transgenic mice models of cerebral ß-amyloidosis are essential to a better understanding of the mechanisms underlying amyloid accumulation in the cerebrovasculature and its interactions with neuritic plaque deposition. Here, our main objective was to evaluate the progression of both parenchymal and vascular deposition in APP23 and 5xFAD transgenic mice in relation to age and sex. We first showed a significant age-dependent accumulation of extracellular Aß deposits in both transgenic models, with a greater increase in APP23 females. We confirmed that CAA pathology was more prominent in the APP23 mice, demonstrating a higher progression of Aß-positive vessels with age, but not linked to sex, and detecting a pronounced burden of cerebral microbleeds (cMBs) by magnetic resonance imaging (MRI). In contrast, 5xFAD mice did not present CAA, as shown by the negligible Aß presence in cerebral vessels and the occurrence of occasional cMBs comparable to WT mice. In conclusion, the APP23 mouse model is an interesting tool to study the overlap between vascular and parenchymal Aß deposition and to evaluate future disease-modifying therapy before its translation to the clinic.


Asunto(s)
Enfermedad de Alzheimer , Amiloidosis , Angiopatía Amiloide Cerebral , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Amiloidosis/patología , Animales , Encéfalo/metabolismo , Angiopatía Amiloide Cerebral/genética , Angiopatía Amiloide Cerebral/patología , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Transgénicos , Placa Amiloide/patología
2.
Circ Res ; 124(1): 114-120, 2019 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-30582445

RESUMEN

RATIONALE: Ischemic stroke is among the leading causes of adult disability. Part of the variability in functional outcome after stroke has been attributed to genetic factors but no locus has been consistently associated with stroke outcome. OBJECTIVE: Our aim was to identify genetic loci influencing the recovery process using accurate phenotyping to produce the largest GWAS (genome-wide association study) in ischemic stroke recovery to date. METHODS AND RESULTS: A 12-cohort, 2-phase (discovery-replication and joint) meta-analysis of GWAS included anterior-territory and previously independent ischemic stroke cases. Functional outcome was recorded using 3-month modified Rankin Scale. Analyses were adjusted for confounders such as discharge National Institutes of Health Stroke Scale. A gene-based burden test was performed. The discovery phase (n=1225) was followed by open (n=2482) and stringent joint-analyses (n=1791). Those cohorts with modified Rankin Scale recorded at time points other than 3-month or incomplete data on previous functional status were excluded in the stringent analyses. Novel variants in PATJ (Pals1-associated tight junction) gene were associated with worse functional outcome at 3-month after stroke. The top variant was rs76221407 (G allele, ß=0.40, P=1.70×10-9). CONCLUSIONS: Our results identify a set of common variants in PATJ gene associated with 3-month functional outcome at genome-wide significance level. Future studies should examine the role of PATJ in stroke recovery and consider stringent phenotyping to enrich the information captured to unveil additional stroke outcome loci.


Asunto(s)
Isquemia Encefálica/genética , Polimorfismo de Nucleótido Simple , Accidente Cerebrovascular/genética , Proteínas de Uniones Estrechas/genética , Isquemia Encefálica/diagnóstico , Isquemia Encefálica/fisiopatología , Isquemia Encefálica/rehabilitación , Evaluación de la Discapacidad , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Fenotipo , Recuperación de la Función , Factores de Riesgo , Accidente Cerebrovascular/diagnóstico , Accidente Cerebrovascular/fisiopatología , Accidente Cerebrovascular/terapia , Rehabilitación de Accidente Cerebrovascular , Resultado del Tratamiento
3.
Int J Mol Sci ; 22(7)2021 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-33805974

RESUMEN

The semicarbazide-sensitive amine oxidase (SSAO), also known as vascular adhesion protein-1 (VAP-1) or primary amine oxidase (PrAO), is a deaminating enzyme highly expressed in vessels that generates harmful products as a result of its enzymatic activity. As a multifunctional enzyme, it is also involved in inflammation through its ability to bind and promote the transmigration of circulating leukocytes into inflamed tissues. Inflammation is present in different systemic and cerebral diseases, including stroke and Alzheimer's disease (AD). These pathologies show important affectations on cerebral vessels, together with increased SSAO levels. This review summarizes the main roles of SSAO/VAP-1 in human physiology and pathophysiology and discusses the mechanisms by which it can affect the onset and progression of both stroke and AD. As there is an evident interrelationship between stroke and AD, basically through the vascular system dysfunction, the possibility that SSAO/VAP-1 could be involved in the transition between these two pathologies is suggested. Hence, its inhibition is proposed to be an interesting therapeutical approach to the brain damage induced in these both cerebral pathologies.


Asunto(s)
Enfermedad de Alzheimer/terapia , Amina Oxidasa (conteniendo Cobre)/metabolismo , Moléculas de Adhesión Celular/metabolismo , Trastornos Cerebrovasculares/terapia , Accidente Cerebrovascular/terapia , Enfermedad de Alzheimer/metabolismo , Aminas/metabolismo , Animales , Adhesión Celular , Angiopatía Amiloide Cerebral/metabolismo , Trastornos Cerebrovasculares/metabolismo , Progresión de la Enfermedad , Células Endoteliales/metabolismo , Glucosa/metabolismo , Humanos , Inflamación/terapia , Leucocitos/citología , Ratones , Ratas , Accidente Cerebrovascular/metabolismo
4.
Cell Mol Life Sci ; 76(16): 3117-3140, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31165904

RESUMEN

Stroke is a leading cause of death and disability worldwide. However, after years of in-depth research, the pathophysiology of stroke is still not fully understood. Increasing evidence shows that matrix metalloproteinases (MMPs) and "a disintegrin and metalloproteinase" (ADAMs) participate in the neuro-inflammatory cascade that is triggered during stroke but also in recovery phases of the disease. This review covers the involvement of these proteins in brain injury following cerebral ischemia which has been widely studied in recent years, with efforts to modulate this group of proteins in neuroprotective therapies, together with their implication in neurorepair mechanisms. Moreover, the review also discusses the role of these proteins in specific forms of neurovascular disease, such as small vessel diseases and intracerebral hemorrhage. Finally, the potential use of MMPs and ADAMs as guiding biomarkers of brain injury and repair for decision-making in cases of stroke is also discussed.


Asunto(s)
Proteínas ADAM/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Accidente Cerebrovascular/patología , Biomarcadores/sangre , Barrera Hematoencefálica/metabolismo , Enfermedades de los Pequeños Vasos Cerebrales/metabolismo , Enfermedades de los Pequeños Vasos Cerebrales/patología , Humanos , Hemorragias Intracraneales/metabolismo , Hemorragias Intracraneales/patología , Accidente Cerebrovascular/metabolismo
5.
Biochim Biophys Acta Mol Basis Dis ; 1864(2): 542-553, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29175057

RESUMEN

Beyond cholesterol reduction, statins mediate their beneficial effects on stroke patients through pleiotropic actions. They have shown anti-inflammatory properties by a number of different mechanisms, including the inhibition of NF-κB transcriptional activity and the consequent increase and release of adhesion molecules. We have studied simvastatin's effects on the vascular enzyme semicarbazide-sensitive amine oxidase/vascular adhesion protein 1 (SSAO/VAP-1), which is involved in stroke-mediated brain injury. SSAO/VAP-1 has leukocyte-binding capacity and mediates the expression of other adhesion proteins through signaling molecules generated by its catalytic activity. Our results indicate that soluble SSAO/VAP-1 is released into the bloodstream after an ischemic stimulus, in parallel with an increase in E-selectin and VCAM-1 and correlating with infarct volume. Simvastatin blocks soluble SSAO/VAP-1 release and prevents E-selectin and VCAM-1 overexpression as well. Simvastatin also effectively blocks SSAO/VAP-1-mediated leukocyte adhesion, although it is not an enzymatic inhibitor of SSAO in vitro. In addition, simvastatin-induced changes in adhesion molecules are greater in human brain endothelial cell cultures expressing SSAO/VAP-1, compared to those not expressing it, indicating some synergic effect with SSAO/VAP-1. We think that part of the beneficial effect of simvastatin in stroke is mediated by the attenuation of the SSAO/VAP-1-dependent inflammatory response.


Asunto(s)
Amina Oxidasa (conteniendo Cobre)/metabolismo , Isquemia Encefálica/metabolismo , Moléculas de Adhesión Celular/metabolismo , Inflamación/metabolismo , Simvastatina/farmacología , Accidente Cerebrovascular/metabolismo , Animales , Encéfalo/metabolismo , Isquemia Encefálica/tratamiento farmacológico , Adhesión Celular , Línea Celular , Modelos Animales de Enfermedad , Selectina E/metabolismo , Células Endoteliales , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Inflamación/tratamiento farmacológico , Masculino , Inhibidor NF-kappaB alfa/metabolismo , FN-kappa B/metabolismo , Ratas , Ratas Wistar , Accidente Cerebrovascular/patología , Molécula 1 de Adhesión Celular Vascular/metabolismo
6.
J Biol Chem ; 290(24): 15078-91, 2015 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-25897080

RESUMEN

Matrix metalloproteases (MMPs) MMP-2 and MMP-9 have been implicated in the physiological catabolism of Alzheimer's amyloid-ß (Aß). Conversely, their association with vascular amyloid deposits, blood-brain barrier disruption, and hemorrhagic transformations after ischemic stroke also highlights their involvement in pathological processes. To better understand this dichotomy, recombinant human (rh) MMP-2 and MMP-9 were incubated with Aß40 and Aß42, and the resulting proteolytic fragments were assessed via immunoprecipitation and quantitative mass spectrometry. Both MMPs generated Aß fragments truncated only at the C terminus, ending at positions 34, 30, and 16. Using deuterated homologues as internal standards, we observed limited and relatively slow degradation of Aß42 by rhMMP-2, although the enzyme cleaved >80% of Aß40 during the 1st h of incubation. rhMMP-9 was significantly less effective, particularly in degrading Aß(1-42), although the targeted peptide bonds were identical. Using Aß(1-34) and Aß(1-30), we demonstrated that these peptides are also substrates for both MMPs, cleaving Aß(1-34) to produce Aß(1-30) first and Aß(1-16) subsequently. Consistent with the kinetics observed with full-length Aß, rhMMP-9 degraded only a minute fraction of Aß(1-34) and was even less effective in producing Aß(1-16). Further degradation of Aß(1-16) by either MMP-2 or MMP-9 was not observed even after prolonged incubation times. Notably, all MMP-generated C-terminally truncated Aß fragments were highly soluble and did not exhibit fibrillogenic properties or induce cytotoxicity in human cerebral microvascular endothelial or neuronal cells supporting the notion that these truncated Aß species are associated with clearance mechanisms rather than being key elements in the fibrillogenesis process.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Western Blotting , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Humanos , Proteolisis , Proteínas Recombinantes/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
7.
J Neurochem ; 136(2): 416-24, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26526443

RESUMEN

A rapid differentiation of acute ischemic stroke and intracerebral hemorrhage (ICH) is essential for an adequate treatment and to promote a better outcome. Our aim was to identify new plasma biomarkers to differentiate stroke subtypes and to combine their diagnostic ability with other biomarkers already described for this clinical indication. Plasma samples of ischemic stroke patients (36) and ICH patients (10) were screened using a 177 antibodies library, and 11 showed different concentrations among stroke subtypes (p < 0.05), mainly chemokines, growth factors and angiogenic factors. Five proteins were selected for replication in 16 ischemic stroke patients and 16 ICH patients, and retinol-binding protein 4 (RPB4), apolipoprotein B100 and pigment epithelial-derived factor were replicated (p < 0.05). These proteins, together with glial fibrillary acidic protein (GFAP) and receptor for advanced glycation end product, were tested in 38 ischemic stroke and 28 ICH samples. Finally, RBP4 >61 µg/mL and GFAP <0.07 ng/mL showed a specificity of 100% for both subtypes. Moreover, after multivariate logistic regression analysis, RBP4 >48.75 µg/mL (ORadj : 6.09 (1.3-28.57), p = 0.02) and GFAP <0.07 ng/mL (ORadj : 0.03 (0.003-0.31), p = 0.003) resulted in independent predictors of stroke subtype, improving discrimination by 29% (p < 0.0001). Both biomarkers might be useful as diagnostic biomarkers to differentiate ischemic stroke and ICH. A rapid differentiation of ischemic stroke from intracerebral hemorrhage is essential to provide the appropriate treatment. We describe the discovery and subsequent replications of RBP4 and its combination with circulating GFAP as plasmatic biomarkers for hyperacute stroke subtype differentiation. The combination of these biomarkers and others might aid to speed up the discrimination of both stroke subtypes improving the outcome of patients.


Asunto(s)
Hemorragia Cerebral/sangre , Proteína Ácida Fibrilar de la Glía/sangre , Proteínas Plasmáticas de Unión al Retinol/metabolismo , Accidente Cerebrovascular/sangre , Anciano , Anciano de 80 o más Años , Isquemia Encefálica/complicaciones , Femenino , Humanos , Masculino , Persona de Mediana Edad , Proteoma/metabolismo , Estudios Retrospectivos , Estadísticas no Paramétricas , Accidente Cerebrovascular/etiología
8.
J Stroke Cerebrovasc Dis ; 25(3): 665-71, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26738811

RESUMEN

BACKGROUND: Intracerebral hemorrhage (ICH) is a devastating form of stroke and depending on the underlying cause, primary ICH is mainly caused by hypertension (HTN-ICH) or cerebral amyloid angiopathy (CAA-ICH). Currently, neuroimaging markers are required to identify the pattern for each etiology. The discovery of new biomarkers to improve the management of this pathology is therefore needed. METHODS: A microarray analysis was carried out to analyze gene expression differences in blood samples from patients (>1.5 months since the last ICH event) who suffered a CAA-ICH and HTN-ICH, and controls. The results were replicated by quantitative polymerase chain reaction and the plasma protein level of the best candidate was measured with enzyme-linked immunosorbent assay. RESULTS: The microarray analysis and the validation study revealed an increase in Golgin A8 Family, Member A (GOLGA8A) mRNA and protein levels in ICH cases compared to controls (P < .01), although no differences were found between specific ICH etiologies. GOLGA8A plasma levels were also associated with the presence of multiple hemorrhages (P < .05). CONCLUSIONS: The GOLGA8A level was increased in the blood of patients who suffered a primary ICH. We did not, however, find any candidate biomarker that distinguished CAA-ICH from HTN-ICH. The role of GOLGA8A in this fatal disorder has yet to be determined.


Asunto(s)
Biomarcadores/sangre , Hemorragia Cerebral/sangre , Hemorragia Cerebral/clasificación , Proteínas de la Membrana/sangre , Análisis por Micromatrices/métodos , Anciano , Angiopatía Amiloide Cerebral/complicaciones , Hemorragia Cerebral/diagnóstico por imagen , Hemorragia Cerebral/etiología , Angiografía por Tomografía Computarizada , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Hipertensión/complicaciones , Procesamiento de Imagen Asistido por Computador , Imagen por Resonancia Magnética , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Persona de Mediana Edad , Estudios Retrospectivos
9.
Stroke ; 46(2): 477-84, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25503547

RESUMEN

BACKGROUND AND PURPOSE: Despite the effectiveness of recombinant tissue-type plasminogen activator (r-tPA) during the acute phase of ischemic stroke, the therapy remains limited by a narrow time window and the occurrence of occasional vascular side effects, particularly symptomatic hemorrhages. Our aim was to investigate the mechanisms underlying the endothelial damage resulting from r-tPA treatment in ischemic-like conditions. METHODS: Microarray analyses were performed on cerebral endothelial cells submitted to r-tPA treatment during oxygen and glucose deprivation to identify novel biomarker candidates. Validation was then performed in vivo in a mouse model of thromboembolic stroke and culminated in an analysis in a clinical cohort of patients with ischemic stroke treated with thrombolysis. RESULTS: The transcription factor NURR1 (NR4A2) was identified as a downstream target induced by r-tPA during oxygen and glucose deprivation. Silencing NURR1 expression reversed the endothelial-toxicity induced by the combined stimuli, a protective effect attributable to reduced levels of proinflammatory mediators, such as nuclear factor-kappa-beta 2 (NF-κ-B2), interleukin 1 alpha (IL1α), intercellular adhesion molecule 1 (ICAM1), SMAD family member 3 (SMAD3), colony stimulating factor 2 (granulocyte-macrophage; CSF2). The detrimental effect of delayed thrombolysis, in conditions in which NURR1 gene expression was enhanced, was confirmed in the preclinical stroke model. Finally, we determined that patients with stroke who had a symptomatic hemorrhagic transformation after r-tPA treatment exhibited higher baseline serum NURR1 levels than did patients with an asymptomatic or absence of cerebral bleedings. CONCLUSIONS: Our results suggest that NURR1 upregulation by r-tPA during ischemic stroke is associated with endothelial dysfunction and inflammation and the enhancement of hemorrhagic complications associated to thrombolysis.


Asunto(s)
Isquemia Encefálica/sangre , Isquemia Encefálica/tratamiento farmacológico , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/sangre , Accidente Cerebrovascular/sangre , Accidente Cerebrovascular/tratamiento farmacológico , Activador de Tejido Plasminógeno/uso terapéutico , Anciano , Anciano de 80 o más Años , Animales , Biomarcadores/sangre , Isquemia Encefálica/diagnóstico , Línea Celular , Femenino , Fibrinolíticos/efectos adversos , Fibrinolíticos/uso terapéutico , Humanos , Inflamación/sangre , Inflamación/inducido químicamente , Inflamación/diagnóstico , Masculino , Ratones , Persona de Mediana Edad , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/biosíntesis , Accidente Cerebrovascular/diagnóstico , Activador de Tejido Plasminógeno/efectos adversos , Resultado del Tratamiento
10.
Stroke ; 45(2): 432-7, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24335228

RESUMEN

BACKGROUND AND PURPOSE: Fluorescent molecular peroxidation products (FMPPs) are considered potential markers of molecular oxidative damage and may provoke increased permeability and disruption of the blood-brain barrier. This study aimed to determine the value of FMPPs as a biomarker to predict neurological worsening related to early hemorrhagic transformation. METHODS: Baseline FMPP levels were measured in 186 consecutive acute ischemic stroke patients before tissue plasminogen activator treatment was administered. A serial FMPP profile (baseline before tissue plasminogen activator treatment, and 1, 2, 12, and 24 hours from treatment) was determined in a subset of 100 patients. Computed tomographic scans were performed at admission and repeated at 24 to 48 hours or after neurological worsening occurred. Symptomatic intracranial hemorrhage was defined as blood at any site in the brain associated with neurological deterioration. RESULTS: Patients who worsened had higher median FMPP levels compared with those who did not (59.68 [48.63-85.73] versus 44.87 [36.37-58.90] Uf/mL; P=0.035) at baseline. After logistic regression multivariate analysis, FMPP >48.2 Uf/mL together with age, hypertension, and systolic blood pressure remained baseline predictors of worsening at 48 hours. Moreover, baseline FMPP determination helped to distinguish between patients who worsened and those who did not (Integrated Discrimination Improvement index, 5.7%; P=0.0004). Finally, within patients who had worsened at 48 hours, those with symptomatic intracranial hemorrhage had higher FMPP levels (P=0.038). CONCLUSIONS: FMPPs might be a valuable biomarker of poor early neurological outcome and be related to the appearance of symptomatic intracranial hemorrhage in tissue plasminogen activator-treated patients, one of the most feared neurological complications after thrombolytic treatment of acute ischemic stroke.


Asunto(s)
Biomarcadores/sangre , Hemorragia Cerebral/etiología , Enfermedades del Sistema Nervioso/etiología , Peróxidos/sangre , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/tratamiento farmacológico , Terapia Trombolítica/efectos adversos , Anciano , Análisis de Varianza , Hemorragia Cerebral/diagnóstico por imagen , Hemorragia Cerebral/epidemiología , Progresión de la Enfermedad , Determinación de Punto Final , Femenino , Fibrinolíticos/efectos adversos , Fibrinolíticos/uso terapéutico , Fluorescencia , Humanos , Masculino , Estrés Oxidativo , Pronóstico , Activador de Tejido Plasminógeno/efectos adversos , Activador de Tejido Plasminógeno/uso terapéutico , Tomografía Computarizada por Rayos X
11.
J Neurochem ; 130(2): 301-12, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24661059

RESUMEN

Finding an efficient neuroprotectant is of urgent need in the field of stroke research. The goal of this study was to test the effect of acute simvastatin administration after stroke in a rat embolic model and to explore its mechanism of action through brain proteomics. To that end, male Wistar rats were subjected to a Middle Cerebral Arteria Occlusion and simvastatin (20 mg/kg s.c) (n = 11) or vehicle (n = 9) were administered 15 min after. To evaluate the neuroprotective mechanisms of simvastatin, brain homogenates after 48 h were analyzed by two-dimensional fluorescence Difference in Gel Electrophoresis (DIGE) technology. We confirmed that simvastatin reduced the infarct volume and improved neurological impairment at 48 h after the stroke in this model. Considering our proteomics analysis, 66 spots, which revealed significant differences between groups, were analyzed by matrix-assisted laser desorption/ionization-time of flight mass spectrometry allowing the identification of 27 proteins. From these results, we suggest that simvastatin protective effect can be partly explained by the attenuation of the oxidative and stress response at blood-brain barrier level after cerebral ischemia. Interestingly, analyzing one of the proteins (HSP75) in plasma from stroke patients who had received simvastatin during the acute phase, we confirmed the results found in the pre-clinical model. Our aim was to study statins benefits when administered during the acute phase of stroke and to explore its mechanisms of action through brain proteomics assay. Using an embolic model, simvastatin-treated rats showed significant infarct volume reduction and neurological improvement compared to vehicle-treated group. Analyzing their homogenated brains by two-dimensional fluorescence Difference in Gel Electrophoresis (DIGE) technology, we concluded that the protective effect of simvastatin can be attributable to oxidative stress response attenuation and blood-brain barrier protection after cerebral ischemia.


Asunto(s)
Química Encefálica/genética , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Embolia Intracraneal/tratamiento farmacológico , Proteómica/métodos , Simvastatina/farmacología , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Western Blotting , Encéfalo/patología , Electroforesis en Gel Bidimensional , Proteínas HSP90 de Choque Térmico/sangre , Humanos , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/patología , Embolia Intracraneal/mortalidad , Embolia Intracraneal/patología , Masculino , Examen Neurológico , Ratas , Ratas Wistar , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Accidente Cerebrovascular/mortalidad , Accidente Cerebrovascular/patología
12.
J Neural Transm (Vienna) ; 121(2): 113-7, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24036696

RESUMEN

Most of neuroprotective strategies in stroke have failed to move from bench to bedside. One explanation might be the use of excessive uniform and smooth experimental models. Therefore, we have employed a more stringent in vitro model based on cultured brain slices from adult mice submitted to OGD. Using this acute model, we have confirmed that mild hypothermia protects against OGD-induced cell death when cooling the tissue during and after OGD, but not when hypothermia is induced only during reoxygenation.


Asunto(s)
Encéfalo/citología , Glucosa/deficiencia , Hipotermia Inducida/métodos , Hipoxia , Neuronas/fisiología , Animales , Muerte Celular/fisiología , Supervivencia Celular/fisiología , L-Lactato Deshidrogenasa/metabolismo , Ratones , Técnicas de Cultivo de Órganos , Estadísticas no Paramétricas
13.
Biomed Pharmacother ; 175: 116779, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38776681

RESUMEN

Diabetic patients present increased volume and functional alterations in epicardial adipose tissue (EAT). We aimed to analyze EAT from type 2 diabetic patients and the inflammatory and cytotoxic effects induced on cardiomyocytes. Furthermore, we analyzed the cardioprotective role of apolipoprotein J (apoJ). EAT explants were obtained from nondiabetic patients (ND), diabetic patients without coronary disease (DM), and DM patients with coronary disease (DM-C) after heart surgery. Morphological characteristics and gene expression were evaluated. Explants were cultured for 24 h and the content of nonesterified fatty acids (NEFA) and sphingolipid species in secretomes was evaluated by lipidomic analysis. Afterwards, secretomes were added to AC16 human cardiomyocytes for 24 h in the presence or absence of cardioprotective molecules (apoJ and HDL). Cytokine release and apoptosis/necrosis were assessed by ELISA and flow cytometry. The EAT from the diabetic samples showed altered expression of genes related to lipid accumulation, insulin resistance, and inflammation. The secretomes from the DM samples presented an increased ratio of pro/antiatherogenic ceramide (Cer) species, while those from DM-C contained the highest concentration of saturated NEFA. DM and DM-C secretomes promoted inflammation and cytotoxicity on AC16 cardiomyocytes. Exogenous Cer16:0, Cer24:1, and palmitic acid reproduced deleterious effects in AC16 cells. These effects were attenuated by exogenous apoJ. Diabetic secretomes promoted inflammation and cytotoxicity in cardiomyocytes. This effect was exacerbated in the secretomes of the DM-C samples. The increased content of specific NEFA and ceramide species seems to play a key role in inducing such deleterious effects, which are attenuated by apoJ.


Asunto(s)
Tejido Adiposo , Diabetes Mellitus Tipo 2 , Inflamación , Miocitos Cardíacos , Pericardio , Humanos , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Tejido Adiposo/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Pericardio/metabolismo , Pericardio/patología , Diabetes Mellitus Tipo 2/metabolismo , Inflamación/patología , Inflamación/metabolismo , Masculino , Femenino , Persona de Mediana Edad , Anciano , Apoptosis/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Ácidos Grasos no Esterificados/metabolismo , Ácidos Grasos no Esterificados/farmacología , Tejido Adiposo Epicárdico
14.
Neurology ; 102(9): e209244, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38598746

RESUMEN

BACKGROUND AND OBJECTIVES: The time taken to achieve blood pressure (BP) control could be pivotal in the benefits of reducing BP in acute intracerebral hemorrhage (ICH). We aimed to assess the relationship between the rapid achievement and sustained maintenance of an intensive systolic BP (SBP) target with radiologic, clinical, and functional outcomes. METHODS: Rapid, Intensive, and Sustained BP lowering in Acute ICH (RAINS) was a multicenter, prospective, observational cohort study of adult patients with ICH <6 hours and SBP ≥150 mm Hg at 4 Comprehensive Stroke Centers during a 4.5-year period. Patients underwent baseline and 24-hour CT scans and 24-hour noninvasive BP monitoring. BP was managed under a rapid (target achievement ≤60 minutes), intensive (target SBP <140 mm Hg), and sustained (target stability for 24 hours) BP protocol. SBP target achievement ≤60 minutes and 24-hour SBP variability were recorded. Outcomes included hematoma expansion (>6 mL or >33%) at 24 hours (primary outcome), early neurologic deterioration (END, 24-hour increase in NIH Stroke Scale score ≥4), and 90-day ordinal modified Rankin scale (mRS) score. Analyses were adjusted by age, sex, anticoagulation, onset-to-imaging time, ICH volume, and intraventricular extension. RESULTS: We included 312 patients (mean age 70.2 ± 13.3 years, 202 [64.7%] male). Hematoma expansion occurred in 70/274 (25.6%) patients, END in 58/291 (19.9%), and the median 90-day mRS score was 4 (interquartile range, 2-5). SBP target achievement ≤60 minutes (178/312 [57.1%]) associated with a lower risk of hematoma expansion (adjusted odds ratio [aOR] 0.43, 95% confidence interval [CI] 0.23-0.77), lower END rate (aOR 0.43, 95% CI 0.23-0.80), and lower 90-day mRS scores (aOR 0.48, 95% CI 0.32-0.74). The mean 24-hour SBP variability was 21.0 ± 7.6 mm Hg. Higher 24-hour SBP variability was not related to expansion (aOR 0.99, 95% CI 0.95-1.04) but associated with higher END rate (aOR 1.15, 95% CI 1.09-1.21) and 90-day mRS scores (aOR 1.06, 95% CI 1.04-1.10). DISCUSSION: Among patients with acute ICH, achieving an intensive SBP target within 60 minutes was associated with lower hematoma expansion risk. Rapid SBP reduction and stable sustention within 24 hours were related to improved clinical and functional outcomes. These findings warrant the design of randomized clinical trials examining the impact of effectively achieving rapid, intensive, and sustained BP control on hematoma expansion. CLASSIFICATION OF EVIDENCE: This study provides Class III evidence that in adults with spontaneous ICH and initial SBP ≥150 mm Hg, lowering SBP to <140 mm Hg within the first hour and maintaining this for 24 hours is associated with decreased hematoma expansion.


Asunto(s)
Hipotensión , Accidente Cerebrovascular , Adulto , Humanos , Masculino , Persona de Mediana Edad , Anciano , Anciano de 80 o más Años , Femenino , Presión Sanguínea/fisiología , Antihipertensivos/uso terapéutico , Antihipertensivos/farmacología , Estudios Prospectivos , Hemorragia Cerebral/diagnóstico por imagen , Hemorragia Cerebral/tratamiento farmacológico , Accidente Cerebrovascular/tratamiento farmacológico , Hematoma/diagnóstico por imagen , Hematoma/tratamiento farmacológico , Resultado del Tratamiento
15.
Cell Death Discov ; 10(1): 85, 2024 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-38368420

RESUMEN

Through GWAS studies we identified PATJ associated with functional outcome after ischemic stroke (IS). The aim of this study was to determine PATJ role in brain endothelial cells (ECs) in the context of stroke outcome. PATJ expression analyses in patient's blood revealed that: (i) the risk allele of rs76221407 induces higher expression of PATJ, (ii) PATJ is downregulated 24 h after IS, and (iii) its expression is significantly lower in those patients with functional independence, measured at 3 months with the modified Rankin scale ((mRS) ≤2), compared to those patients with marked disability (mRS = 4-5). In mice brains, PATJ was also downregulated in the injured hemisphere at 48 h after ischemia. Oxygen-glucose deprivation and hypoxia-dependent of Hypoxia Inducible Factor-1α also caused PATJ depletion in ECs. To study the effects of PATJ downregulation, we generated PATJ-knockdown human microvascular ECs. Their transcriptomic profile evidenced a complex cell reprogramming involving Notch, TGF-ß, PI3K/Akt, and Hippo signaling that translates in morphological and functional changes compatible with endothelial to mesenchymal transition (EndMT). PATJ depletion caused loss of cell-cell adhesion, upregulation of metalloproteases, actin cytoskeleton remodeling, cytoplasmic accumulation of the signal transducer C-terminal transmembrane Mucin 1 (MUC1-C) and downregulation of Notch and Hippo signaling. The EndMT phenotype of PATJ-depleted cells was associated with the nuclear recruitment of MUC1-C, YAP/TAZ, ß-catenin, and ZEB1. Our results suggest that PATJ downregulation 24 h after IS promotes EndMT, an initial step prior to secondary activation of a pro-angiogenic program. This effect is associated with functional independence suggesting that activation of EndMT shortly after stroke onset is beneficial for stroke recovery.

16.
J Cell Mol Med ; 17(12): 1543-53, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23945132

RESUMEN

The enhancement of endogenous angiogenesis after stroke will be critical in neurorepair therapies where endothelial progenitor cells (EPCs) might be key players. Our aim was to determine the influence of cerebral ischaemia and the role of matrix metalloproteinase-9 (MMP-9) on the angiogenic function of EPCs. Permanent focal cerebral ischaemia was induced by middle cerebral artery (MCA) occlusion in MMP-9/knockout (MMP-9/KO) and wild-type (WT) mice. EPCs were obtained for cell counting after ischaemia (6 and 24 hrs) and in control animals. Matrigel(™) assays and time-lapse imaging were conducted to monitor angiogenic function of WT and MMP9-deficient EPCs or after treatment with MMP-9 inhibitors. Focal cerebral ischaemia increased the number of early EPCs, while MMP-9 deficiency decreased their number in non-ischaemic mice and delayed their release after ischaemia. Late outgrowth endothelial cells (OECs) from ischaemic mice shaped more vessel structures than controls, while MMP-9 deficiency reduced the angiogenic abilities of OECs to form vascular networks, in vitro. Treatment with the MMP inhibitor GM6001 and the specific MMP-9 inhibitor I also decreased the number of vessel structures shaped by both human and mouse WT OECs, while exogenous MMP-9 could not revert the impaired angiogenic function in MMP-9/KO OECs. Finally, time-lapse imaging showed that the extension of vascular networks was influenced by cerebral ischaemia and MMP-9 deficiency early during the vascular network formation followed by a dynamic vessel remodelling. We conclude that focal cerebral ischaemia triggers the angiogenic responses of EPCs, while MMP-9 plays a key role in the formation of vascular networks by EPCs.


Asunto(s)
Isquemia Encefálica/enzimología , Isquemia Encefálica/fisiopatología , Células Endoteliales/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Neovascularización Fisiológica , Células Madre/metabolismo , Animales , Isquemia Encefálica/patología , Recuento de Células , Colágeno/metabolismo , Combinación de Medicamentos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Humanos , Inmunofenotipificación , Laminina/metabolismo , Masculino , Metaloproteinasa 9 de la Matriz/deficiencia , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Ratones , Neovascularización Fisiológica/efectos de los fármacos , Proteoglicanos/metabolismo , Células Madre/efectos de los fármacos , Células Madre/patología
17.
Br J Pharmacol ; 180(15): 1999-2017, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-36872299

RESUMEN

BACKGROUND AND PURPOSE: Therapies based on apolipoprotein A-I (ApoA-I), classically tested for cardiovascular diseases, were recently proposed for Alzheimer's disease (AD). Based on a drug reprofiling approach, our objective was to explore the use of a natural variant of ApoA-I form, ApoA-I-Milano (M), as a treatment for AD. ApoA-I-M contains the R173C mutation, and confers protection against atherosclerosis development, although ApoA-I-M carriers exhibit low HDL levels. EXPERIMENTAL APPROACH: Middle-aged (12-month-old) and aged (21-month-old) APP23 mice were intraperitoneally treated for 10 weeks with human recombinant ApoA-I-M (hrApoA-I-M) protein or saline. Pathology progression through behavioural parameters and biochemical determinations was evaluated. KEY RESULTS: In middle-aged group, hrApoA-I-M treatment reduced the anxiety behaviour associated with this AD model. In aged mice, hrApoA-I-M reversed T-Maze performance alterations, a cognitive improvement accompanied by neuronal loss recovery in the dentate gyrus. Aged mice treated with hrApoA-I-M showed lower brain Aß40 soluble levels and elevated Aß40 levels in cerebrospinal fluid, without modifying insoluble brain Aß burden. Interestingly, hrApoA-I-M sub-chronic treatment induced a molecular effect on the cerebrovasculature, increasing occludin expression and ICAM-1 presence, as well as promoting an elevation of plasma soluble RAGE in all hrApoA-I-M-treated mice, drastically decreasing the AGEs/sRAGE ratio, a marker of endothelial damage. CONCLUSION AND IMPLICATIONS: Peripheral hrApoA-I-M treatment shows a beneficial impact on working memory, involving mechanisms related with brain Aß mobilization and modulation of the levels of cerebrovascular markers. Our study shows the potential therapeutic applicability of a safe and non-invasive treatment based on peripheral administration of hrApoA-I-M in AD.


Asunto(s)
Enfermedad de Alzheimer , Persona de Mediana Edad , Ratones , Humanos , Animales , Lactante , Enfermedad de Alzheimer/metabolismo , Ratones Transgénicos , Apolipoproteína A-I/genética , Encéfalo/metabolismo , Mutación , Modelos Animales de Enfermedad , Péptidos beta-Amiloides/metabolismo
18.
Front Aging Neurosci ; 15: 1134399, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37113571

RESUMEN

Introduction: Cerebral amyloid angiopathy (CAA) is characterized by the accumulation of amyloid-ß (Aß) in brain vessels and is a main cause of lobar intracerebral hemorrhage (ICH) in the elderly. CAA is associated with magnetic resonance imaging (MRI) markers of small vessel disease (SVD). Since Aß is also accumulated in Alzheimer's disease (AD) in the brain parenchyma, we aimed to study if several single nucleotide polymorphisms (SNPs) previously associated with AD were also associated with CAA pathology. Furthermore, we also studied the influence of APOE and CLU genetic variants in apolipoprotein E (ApoE) and clusterin/apolipoprotein J (ApoJ) circulating levels and their distribution among lipoproteins. Methods: The study was carried out in a multicentric cohort of 126 patients with lobar ICH and clinical suspicion of CAA. Results: We observed several SNPs associated with CAA neuroimaging MRI markers [cortical superficial siderosis (cSS), enlarged perivascular spaces in the centrum semiovale (CSO-EPVS), lobar cerebral microbleeds (CMB), white matter hyperintensities (WMH), corticosubcortical atrophy and CAA-SVD burden score]. Concretely, ABCA7 (rs3764650), CLU (rs9331896 and rs933188), EPHA1 (rs11767557), and TREML2 (rs3747742) were significantly associated with a CAA-SVD burden score. Regarding circulating levels of apolipoproteins, protective AD SNPs of CLU [rs11136000 (T) and rs9331896 (C)] were significantly associated with higher HDL ApoJ content in the lobar ICH cohort. APOEε2 carriers presented higher plasma and LDL-associated ApoE levels whereas APOEε4 carriers presented lower plasma ApoE levels. Additionally, we observed that lower circulating ApoJ and ApoE levels were significantly associated with CAA-related MRI markers. More specifically, lower LDL-associated ApoJ and plasma and HDL-associated ApoE levels were significantly associated with CSO-EPVS, lower ApoJ content in HDL with brain atrophy and lower ApoE content in LDL with the extent of cSS. Discussion: This study reinforces the relevance of lipid metabolism in CAA and cerebrovascular functionality. We propose that ApoJ and ApoE distribution among lipoproteins may be associated with pathological features related to CAA with higher ApoE and ApoJ levels in HDL possibly enhancing atheroprotective, antioxidative, and anti-inflammatory responses in cerebral ß-amyloidosis.

19.
J Neurochem ; 122(2): 233-43, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22548274

RESUMEN

Protective effects of statins have been well documented for stroke therapy. Here, we used a systematic review and meta-analysis to assess these evidences. We identified 190 studies using statin treatment in stroke animal models by electronic searching. From those, only studies describing ischemic occlusive stroke and reporting data on infarct volume and/or neurological outcome were included in the analysis (41 publications, 1882 animals). The global estimate effect was assessed by Weighted Mean Difference meta-analysis. Statins reduced infarct volume by 25.12% (20.66%-29.58%, P < 0.001) and consistently, induced an improvement on neurological outcome (20.36% (14.17%-26.56%), P < 0.001). Stratified analysis showed that simvastatin had the greatest effect on infarct volume reduction (38.18%) and neurological improvement (22.94%), whereas bigger infarct reduction was observed giving the statin as a pre-treatment (33.5%) compared with post-treatment (16.02%). The use of pentobarbital sodium, the timing of statin administration, the statement of conflict of interest and the type of statin studied were found to be independent factors in the meta-regression, indicating their influence on the results of studies examining statin treatment. In conclusion, this meta-analysis provides further evidences of the efficacy of statins, supporting their potential use for human stroke therapy.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/patología , Interpretación Estadística de Datos , Modelos Animales de Enfermedad , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Infarto de la Arteria Cerebral Media/patología , Ratones , Sesgo de Publicación , Ratas , Análisis de Regresión , Proyectos de Investigación
20.
Cerebrovasc Dis ; 33(1): 55-63, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22133888

RESUMEN

BACKGROUND: Vascular adhesion protein-1 (VAP-1) is a cell surface and circulating enzyme that belongs to the semicarbazide-sensitive amine oxidase (SSAO) family, which oxidatively deaminates primary amines and is implicated in leukocyte extravasation. Our aim was to investigate the alteration of soluble VAP-1/SSAO activity in plasma samples after acute intracerebral hemorrhage (ICH) and its presence in human ICH brain tissue. METHODS: VAP-1/SSAO activity was determined in plasma of 66 ICH patients and 58 healthy controls. In addition, we assessed the expression of VAP-1/SSAO in postmortem brain tissue from hemorrhagic stroke patients by Western blot and immunohistochemistry. RESULTS: We observed significantly higher levels of plasma VAP-1/SSAO activity in patients with ICH compared to matched elderly controls (p = 0.001). Plasma VAP-1/SSAO activity <2.7 pmol/min·mg and baseline ICH volume <17 ml were independent predictors of neurological improvement after 48 h (OR 6.8, 95% CI 1.14-41.67, p = 0.035, and OR 10.64, 95% CI 1.1-100, p = 0.041, respectively), after adjustment for baseline stroke severity. We also found that membrane-bound VAP-1/SSAO levels were lower in the perihematoma region than in the corresponding contralateral brain areas of patients deceased due to ICH (p = 0.024). CONCLUSIONS: Our data demonstrate that plasma VAP-1/SSAO activity is increased in ICH and predicts neurological outcome, suggesting a possible contribution of the soluble protein in secondary brain damage. Furthermore, anti-VAP-1/SSAO strategies might be a promising approach to prevent neurological worsening following ICH.


Asunto(s)
Amina Oxidasa (conteniendo Cobre)/sangre , Encéfalo/enzimología , Moléculas de Adhesión Celular/sangre , Hemorragias Intracraneales/enzimología , Accidente Cerebrovascular/enzimología , Anciano , Anciano de 80 o más Años , Autopsia , Biomarcadores/sangre , Western Blotting , Estudios de Casos y Controles , Distribución de Chi-Cuadrado , Femenino , Humanos , Inmunohistoquímica , Hemorragias Intracraneales/sangre , Hemorragias Intracraneales/mortalidad , Modelos Logísticos , Masculino , Persona de Mediana Edad , Análisis Multivariante , Oportunidad Relativa , Pronóstico , España , Accidente Cerebrovascular/sangre , Accidente Cerebrovascular/mortalidad , Factores de Tiempo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA