Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
2.
Nat Immunol ; 21(5): 513-524, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32284594

RESUMEN

Oxidative stress is a central part of innate immune-induced neurodegeneration. However, the transcriptomic landscape of central nervous system (CNS) innate immune cells contributing to oxidative stress is unknown, and therapies to target their neurotoxic functions are not widely available. Here, we provide the oxidative stress innate immune cell atlas in neuroinflammatory disease and report the discovery of new druggable pathways. Transcriptional profiling of oxidative stress-producing CNS innate immune cells identified a core oxidative stress gene signature coupled to coagulation and glutathione-pathway genes shared between a microglia cluster and infiltrating macrophages. Tox-seq followed by a microglia high-throughput screen and oxidative stress gene network analysis identified the glutathione-regulating compound acivicin, with potent therapeutic effects that decrease oxidative stress and axonal damage in chronic and relapsing multiple sclerosis models. Thus, oxidative stress transcriptomics identified neurotoxic CNS innate immune populations and may enable discovery of selective neuroprotective strategies.


Asunto(s)
Encefalomielitis Autoinmune Experimental/genética , Perfilación de la Expresión Génica/métodos , Microglía/fisiología , Esclerosis Múltiple/genética , Inflamación Neurogénica/genética , Animales , Antioxidantes/uso terapéutico , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Femenino , Redes Reguladoras de Genes , Ensayos Analíticos de Alto Rendimiento , Humanos , Inmunidad Innata , Isoxazoles/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Esclerosis Múltiple/tratamiento farmacológico , Inflamación Neurogénica/tratamiento farmacológico , Estrés Oxidativo , Análisis de Secuencia de ARN , Análisis de la Célula Individual
3.
Am J Pathol ; 191(3): 575-583, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33608067

RESUMEN

Central nervous system (CNS) lymphoma is an extranodal non-Hodgkin B-cell lymphoma characterized by malignant lymph tissue arising in the brain or spinal cord, associated with inflammation and blood-brain barrier (BBB) disruption. Although BBB disruption is known to occur in patients with CNS lymphoma, a direct link between these two has not been shown. Herein, abundant deposition of the blood coagulation protein fibrinogen around B-cell lymphoma was detected in CNS lymphoma patients and in the CNS parenchyma in an orthotopic mouse model. Functional enrichment analysis of unbiased cerebrospinal fluid proteomics of CNS B-cell lymphoma patients showed that coagulation protein networks were highly connected with tumor-associated biological signaling pathways. In vivo two-photon imaging demonstrated that lymphoma growth was associated with BBB disruption, and in vitro experiments identified a role for fibrinogen in promoting lymphoma cell adhesion. Overall, these results identify perivascular lymphoma clustering at sites of fibrinogen deposition, and suggest that fibrinogen may be a target for pharmacologic intervention in metastatic B-cell lymphoma associated with BBB disruption.


Asunto(s)
Adhesión Celular , Neoplasias del Sistema Nervioso Central/patología , Fibrinógeno/metabolismo , Inflamación/patología , Linfocitos/patología , Linfoma de Células B/patología , Animales , Transporte Biológico , Neoplasias del Sistema Nervioso Central/etiología , Neoplasias del Sistema Nervioso Central/metabolismo , Modelos Animales de Enfermedad , Fibrinógeno/genética , Humanos , Inflamación/etiología , Inflamación/metabolismo , Linfocitos/metabolismo , Linfoma de Células B/etiología , Linfoma de Células B/metabolismo , Masculino , Ratones , Ratones Desnudos
4.
Brain ; 144(8): 2291-2301, 2021 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-34426831

RESUMEN

Extrinsic inhibitors at sites of blood-brain barrier disruption and neurovascular damage contribute to remyelination failure in neurological diseases. However, therapies to overcome the extrinsic inhibition of remyelination are not widely available and the dynamics of glial progenitor niche remodelling at sites of neurovascular dysfunction are largely unknown. By integrating in vivo two-photon imaging co-registered with electron microscopy and transcriptomics in chronic neuroinflammatory lesions, we found that oligodendrocyte precursor cells clustered perivascularly at sites of limited remyelination with deposition of fibrinogen, a blood coagulation factor abundantly deposited in multiple sclerosis lesions. By developing a screen (OPC-X-screen) to identify compounds that promote remyelination in the presence of extrinsic inhibitors, we showed that known promyelinating drugs did not rescue the extrinsic inhibition of remyelination by fibrinogen. In contrast, bone morphogenetic protein type I receptor blockade rescued the inhibitory fibrinogen effects and restored a promyelinating progenitor niche by promoting myelinating oligodendrocytes, while suppressing astrocyte cell fate, with potent therapeutic effects in chronic models of multiple sclerosis. Thus, abortive oligodendrocyte precursor cell differentiation by fibrinogen is refractory to known promyelinating compounds, suggesting that blockade of the bone morphogenetic protein signalling pathway may enhance remyelinating efficacy by overcoming extrinsic inhibition in neuroinflammatory lesions with vascular damage.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Receptores de Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Oligodendroglía/efectos de los fármacos , Remielinización/efectos de los fármacos , Médula Espinal/efectos de los fármacos , Animales , Barrera Hematoencefálica/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular/efectos de los fármacos , Homeostasis/efectos de los fármacos , Ratones , Ratones Transgénicos , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/metabolismo , Células Precursoras de Oligodendrocitos/efectos de los fármacos , Células Precursoras de Oligodendrocitos/metabolismo , Oligodendroglía/metabolismo , Pirazoles/farmacología , Pirimidinas/farmacología , Quinolinas/farmacología , Médula Espinal/metabolismo
5.
Eur J Clin Invest ; 51(11): e13600, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34076259

RESUMEN

AIMS: Epidemiologic evidence links ischemic stroke to age, yet the mechanisms that underlie the specific and independent effects of age on stroke remain elusive, impeding the development of targeted treatments. This study tested the hypothesis that age directly aggravates stroke outcomes and proposes inflamm-aging as a mediator and potential therapeutic target. METHODS: 3 months- (young) and 18-20 months-old (old) mice underwent transient middle cerebral artery occlusion (tMCAO) for 30 minutes followed by 48 hours of reperfusion. Old animals received weekly treatment with the TNF-α neutralizing antibody adalimumab over 4 weeks before tMCAO in a separate set of experiments. Plasma levels of TNF- α were assessed in patients with ischemic stroke and correlated with age and outcome. RESULTS: Old mice displayed larger stroke size than young ones with increased neuromotor deficit. Immunohistochemical analysis revealed impairment of the blood-brain barrier in old mice, i.e. increased post-stroke degradation of endothelial tight junctions and expression of tight junctions-digesting and neurotoxic matrix metalloproteinases. At baseline, old animals showed a broad modulation of several circulating inflammatory mediators. TNF-α displayed the highest increase in old animals and its inhibition restored the volume of stroke, neuromotor performance, and survival rates of old mice to the levels observed in young ones. Patients with ischemic stroke showed increased TNF-α plasma levels which correlated with worsened short-term neurological outcome as well as with age. CONCLUSIONS: This study identifies TNF-α as a causative contributor to the deleterious effect of aging on stroke and points to inflamm-aging as a mechanism of age-related worsening of stroke outcomes and potential therapeutic target in this context. Thus, this work provides a basis for tailoring novel stroke therapies for the particularly vulnerable elderly population.


Asunto(s)
Adalimumab/farmacología , Envejecimiento/efectos de los fármacos , Infarto de la Arteria Cerebral Media/metabolismo , Inflamación/metabolismo , Inhibidores del Factor de Necrosis Tumoral/farmacología , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Anciano , Anciano de 80 o más Años , Envejecimiento/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Cadherinas/metabolismo , Femenino , Humanos , Interleucina-1beta/metabolismo , Accidente Cerebrovascular Isquémico/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Recuperación de la Función , Daño por Reperfusión/metabolismo , Proteínas de Uniones Estrechas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
6.
Proc Natl Acad Sci U S A ; 114(19): 5029-5034, 2017 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-28438992

RESUMEN

Frontotemporal dementia (FTD) is the second most common dementia before 65 years of age. Haploinsufficiency in the progranulin (GRN) gene accounts for 10% of all cases of familial FTD. GRN mutation carriers have an increased risk of autoimmune disorders, accompanied by elevated levels of tissue necrosis factor (TNF) α. We examined behavioral alterations related to obsessive-compulsive disorder (OCD) and the role of TNFα and related signaling pathways in FTD patients with GRN mutations and in mice lacking progranulin (PGRN). We found that patients and mice with GRN mutations displayed OCD and self-grooming (an OCD-like behavior in mice), respectively. Furthermore, medium spiny neurons in the nucleus accumbens, an area implicated in development of OCD, display hyperexcitability in PGRN knockout mice. Reducing levels of TNFα in PGRN knockout mice abolished excessive self-grooming and the associated hyperexcitability of medium spiny neurons of the nucleus accumbens. In the brain, PGRN is highly expressed in microglia, which are a major source of TNFα. We therefore deleted PGRN specifically in microglia and found that it was sufficient to induce excessive grooming. Importantly, excessive grooming in these mice was prevented by inactivating nuclear factor κB (NF-κB) in microglia/myeloid cells. Our findings suggest that PGRN deficiency leads to excessive NF-κB activation in microglia and elevated TNFα signaling, which in turn lead to hyperexcitability of medium spiny neurons and OCD-like behavior.


Asunto(s)
Demencia Frontotemporal/metabolismo , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Microglía/metabolismo , FN-kappa B/metabolismo , Trastorno Obsesivo Compulsivo/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Modelos Animales de Enfermedad , Femenino , Demencia Frontotemporal/genética , Demencia Frontotemporal/patología , Granulinas , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Masculino , Ratones , Ratones Noqueados , Microglía/patología , FN-kappa B/genética , Trastorno Obsesivo Compulsivo/genética , Trastorno Obsesivo Compulsivo/patología , Progranulinas , Factor de Necrosis Tumoral alfa/genética
7.
Stroke ; 50(2): 469-477, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30626291

RESUMEN

Background and Purpose- Inflammation is a major pathogenic component of ischemia/reperfusion brain injury, and as such, interventions aimed at inhibiting inflammatory mediators promise to be effective strategies in stroke therapy. JunD-a member of the AP-1 (activated protein-1) family of transcription factors-was recently shown to regulate inflammation by targeting IL (interleukin)-1ß synthesis and macrophage activation. The purpose of the present study was to assess the role of JunD in ischemia/reperfusion-induced brain injury. Methods- WT (wild type) mice randomly treated with either JunD or scramble (control) siRNA were subjected to 45 minutes of transient middle cerebral artery occlusion followed by 24 hours of reperfusion. Stroke size, neurological deficit, plasma/brain cytokines, and oxidative stress determined by 4-hydroxynonenal immunofluorescence staining were evaluated 24 hours after reperfusion. Additionally, the role of IL-1ß was investigated by treating JunD siRNA mice with an anti-IL-1ß monoclonal antibody on reperfusion. Finally, JunD expression was assessed in peripheral blood monocytes isolated from patients with acute ischemic stroke. Results- In vivo JunD knockdown resulted in increased stroke size, reduced neurological function, and increased systemic inflammation, as confirmed by higher neutrophil count and lymphopenia. Brain tissue IL-1ß levels were augmented in JunD siRNA mice as compared with scramble siRNA, whereas no difference was detected in IL-6, TNF-α (tumor necrosis factor-α), and 4-hydroxynonenal levels. The deleterious effects of silencing of JunD were rescued by treating mice with an anti-IL-1ß antibody. In addition, JunD expression was decreased in peripheral blood monocytes of patients with acute ischemic stroke at 6 and 24 hours after onset of stroke symptoms compared with sex- and age-matched healthy controls. Conclusions- JunD blunts ischemia/reperfusion-induced brain injury via suppression of IL-1ß.


Asunto(s)
Lesiones Encefálicas/metabolismo , Interleucina-1beta/metabolismo , Estrés Oxidativo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Daño por Reperfusión/metabolismo , Animales , Lesiones Encefálicas/genética , Lesiones Encefálicas/patología , Regulación de la Expresión Génica , Interleucina-1beta/genética , Masculino , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas c-jun/genética , Daño por Reperfusión/genética , Daño por Reperfusión/patología
8.
J Neurosci ; 37(45): 10808-10816, 2017 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-29118209

RESUMEN

In vivo optical imaging has emerged as a powerful tool with which to study cellular responses to injury and disease in the mammalian CNS. Important new insights have emerged regarding axonal degeneration and regeneration, glial responses and neuroinflammation, changes in the neurovascular unit, and, more recently, neural transplantations. Accompanying a 2017 SfN Mini-Symposium, here, we discuss selected recent advances in understanding the neuronal, glial, and other cellular responses to CNS injury and disease with in vivo imaging of the rodent brain or spinal cord. We anticipate that in vivo optical imaging will continue to be at the forefront of breakthrough discoveries of fundamental mechanisms and therapies for CNS injury and disease.


Asunto(s)
Enfermedades del Sistema Nervioso Central/diagnóstico por imagen , Sistema Nervioso Central/diagnóstico por imagen , Sistema Nervioso Central/lesiones , Neuroimagen/métodos , Animales , Humanos , Ratones , Neuroimagen/instrumentación , Ratas
9.
Neurodegener Dis ; 18(1): 49-56, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29402847

RESUMEN

BACKGROUND: Strong genetic and epidemiological evidence points to a crucial role of the immune system in the development of Alzheimer disease (AD). CD3+ T lymphocytes have been described in brains of postmortem AD patients and in transgenic models of AD-like cerebral amyloidosis and tau pathology. However, the occurrence of T cells in AD brains is still controversial; furthermore, the relationship between T cells and hallmarks of AD pathology (amyloid plaques and neurofibrillary tangles) remains to be established. OBJECTIVES: We have studied the occurrence of T cells in postmortem hippocampi and mid frontal gyrus (MFG) samples of AD patients (Braak stage V-VI) and nondemented control subjects and correlated it with amyloid and tau pathology burden. METHODS: Confocal microscopy and bright-field immunohistochemistry were used to identify brain-associated T cells. Extravascular CD3+ T cells were quantified and compared to nondemented controls. In addition, numbers of extravascular CD3+ T cells were correlated with amyloid (6E10 staining) and tau pathology (AT8 staining) in the same sections. RESULTS: Several CD3+, extravascular T cells were observed in the brains of AD patients, mostly of the CD8+ subtype. AD hippocampi harbored significantly increased numbers of extravascular CD3+ T cells compared to nondemented controls. CD3+ T cells significantly correlated with tau pathology but not with amyloid plaques in AD samples. CONCLUSIONS: Our data support the notion of T-cell occurrence in AD brains and suggest that, in advanced stages of AD, T-cell extravasation is driven by tau-related neurodegenerative changes rather than by cerebral amyloidosis. T cells could be crucial for driving the amyloid-independent phase of the AD pathology.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Encéfalo/inmunología , Complejo CD3/inmunología , Placa Amiloide/inmunología , Linfocitos T/inmunología , Anciano de 80 o más Años , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Encéfalo/metabolismo , Encéfalo/patología , Complejo CD3/metabolismo , Femenino , Humanos , Masculino , Placa Amiloide/metabolismo , Placa Amiloide/patología , Linfocitos T/metabolismo
10.
Am J Physiol Heart Circ Physiol ; 312(2): H232-H238, 2017 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-27836896

RESUMEN

In Alzheimer's disease (AD), cerebral arteries, in contrast to cerebral microvessels, show both cerebral amyloid angiopathy (CAA) -dependent and -independent vessel wall pathology. However, it remains unclear whether CAA-independent vessel wall pathology affects arterial function, thereby chronically reducing cerebral perfusion, and, if so, which mechanisms mediate this effect. To this end, we assessed the ex vivo vascular function of the basilar artery and a similar-sized peripheral artery (femoral artery) in the Swedish-Arctic (SweArc) transgenic AD mouse model at different disease stages. Furthermore, we used quantitative immunohistochemistry to analyze CAA, endothelial morphology, and molecular pathways pertinent to vascular relaxation. We found that endothelium-dependent, but not smooth muscle-dependent, vasorelaxation was significantly impaired in basilar and femoral arteries of 15-mo-old SweArc mice compared with that of age-matched wild-type and 6-mo-old SweArc mice. This impairment was accompanied by significantly reduced levels of cyclic GMP, indicating a reduced nitric oxide (NO) bioavailability. However, no age- and genotype-related differences in oxidative stress as measured by lipid peroxidation were observed. Although parenchymal capillaries, arterioles, and arteries showed abundant CAA in the 15-mo-old SweArc mice, no CAA or changes in endothelial morphology were detected histologically in the basilar and femoral artery. Thus our results suggest that, in this AD mouse model, dysfunction of large intracranial, extracerebral arteries important for brain perfusion is mediated by reduced NO bioavailability rather than by CAA. This finding supports the growing body of evidence highlighting the therapeutic importance of targeting the cerebrovasculature in AD. NEW & NOTEWORTHY: We show that vasorelaxation of the basilar artery, a large intracranial, extracerebral artery important for cerebral perfusion, is impaired independent of cerebral amyloid angiopathy in a transgenic mouse model of Alzheimer's disease. Interestingly, this dysfunction is specifically endothelium related and is mediated by impaired nitric oxide-cyclic GMP bioavailability.


Asunto(s)
Envejecimiento/metabolismo , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Arteria Basilar/metabolismo , Angiopatía Amiloide Cerebral/metabolismo , Óxido Nítrico/metabolismo , Envejecimiento/fisiología , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Animales , Arteria Basilar/patología , Arteria Basilar/fisiopatología , Disponibilidad Biológica , Angiopatía Amiloide Cerebral/patología , Angiopatía Amiloide Cerebral/fisiopatología , Arterias Cerebrales/metabolismo , Arterias Cerebrales/patología , Arterias Cerebrales/fisiopatología , Circulación Cerebrovascular/fisiología , GMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Endotelio Vascular/fisiopatología , Arteria Femoral/metabolismo , Arteria Femoral/patología , Arteria Femoral/fisiopatología , Inmunohistoquímica , Masculino , Ratones , Ratones Transgénicos , Vasodilatación/fisiología
11.
Vasc Med ; 22(4): 285-291, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28643554

RESUMEN

Arterial distensibility, an independent predictor of cardiovascular events, is transiently increased with acute hyperglycemia (AHG) in healthy individuals. Whether this response interacts with physical inactivity remains unknown. We examined the effects of short-term bed rest (BR) on the response of carotid artery distensibility (CD) to AHG, and the influence of underlying changes in insulin resistance and blood volume. CD was assessed with ultrasonography before as well as 30 and 120 minutes following ingestion of 75 g of glucose prior to and after 3 days of BR in 15 healthy male volunteers. Plasma insulin/glucose concentrations and blood volumes were concomitantly determined. On day 4 of BR, blood volume was re-established to pre-BR levels by albumin infusion and CD and insulin/glucose concentrations were determined as in the previous experimental days. Basal CD was not affected by BR. AHG increased CD before and after BR but reached a higher peak increment after BR (12% vs 60% at 30 min OGTT, p=0.028). BR also increased the plasma insulin concentration during AHG ( p=0.007). In regression analyses, plasma insulin and glucose concentrations were positively correlated to CD, particularly after BR ( r=0.31, p<0.05). Restoration of the BR-induced loss (5%) in blood volume did not affect the response of CD to AHG. In conclusion, short-term physical inactivity strongly accentuates the initial increase in CD in response to AHG in healthy individuals. This effect is associated with concomitant increases in circulating insulin concentration attributable to early insulin resistance.


Asunto(s)
Reposo en Cama/efectos adversos , Glucemia/metabolismo , Enfermedades Cardiovasculares/etiología , Arterias Carótidas/fisiopatología , Ejercicio Físico , Hiperglucemia/complicaciones , Rigidez Vascular , Enfermedad Aguda , Adulto , Biomarcadores/sangre , Volumen Sanguíneo , Enfermedades Cardiovasculares/fisiopatología , Arterias Carótidas/diagnóstico por imagen , Prueba de Tolerancia a la Glucosa , Voluntarios Sanos , Hemodinámica , Humanos , Hiperglucemia/sangre , Hiperglucemia/diagnóstico , Insulina/sangre , Resistencia a la Insulina , Masculino , Factores de Riesgo , Factores de Tiempo , Ultrasonografía , Adulto Joven
12.
Acta Neuropathol ; 131(5): 737-52, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26988843

RESUMEN

Alzheimer's disease (AD) is characterised by pathologic cerebrovascular remodelling. Whether this occurs already before disease onset, as may be indicated by early Braak tau-related cerebral hypoperfusion and blood-brain barrier (BBB) impairment found in previous studies, remains unknown. Therefore, we systematically quantified Braak tau stage- and cerebral amyloid angiopathy (CAA)-dependent alterations in the alpha-smooth muscle actin (α-SMA), collagen, and elastin content of leptomeningeal arterioles, small arteries, and medium-sized arteries surrounding the gyrus frontalis medialis (GFM) and hippocampus (HIPP), including the sulci, of 17 clinically and pathologically diagnosed AD subjects (Braak stage IV-VI) and 28 non-demented control subjects (Braak stage I-IV). GFM and HIPP paraffin sections were stained for general collagen and elastin with the Verhoeff-van Gieson stain; α-SMA and CAA/amyloid ß (Aß) were detected using immunohistochemistry. Significant arterial elastin degradation was observed from Braak stage III onward and correlated with Braak tau pathology (ρ = 0.909, 95% CI 0.370 to 0.990, p < 0.05). This was accompanied by an increase in neutrophil elastase expression by α-SMA-positive cells in the vessel wall. Small and medium-sized arteries exhibited significant CAA-independent α-SMA loss starting between Braak stage I and II-III, along with accumulation of phosphorylated paired helical filament (PHF) tau in the perivascular space of intraparenchymal vessels. α-SMA remained at the decreased level throughout the later Braak stages. In contrast, arterioles exhibited significant α-SMA loss only at Braak stage V and VI/in AD subjects, which was CAA-dependent/correlated with CAA burden (ρ = -0.422, 95% CI -0.557 to -0.265, p < 0.0001). Collagen content was only significantly changed in small arteries. Our data indicate that vessel wall remodelling of leptomeningeal arteries is an early-onset, Braak tau pathology-dependent process unrelated to CAA and AD, which potentially may contribute to downstream CAA-dependent microvascular pathology in AD.


Asunto(s)
Enfermedad de Alzheimer/patología , Encéfalo/metabolismo , Encéfalo/patología , Angiopatía Amiloide Cerebral/patología , Microvasos/patología , Proteínas tau/metabolismo , Actinas/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/diagnóstico por imagen , Análisis de Varianza , Barrera Hematoencefálica/patología , Angiopatía Amiloide Cerebral/diagnóstico por imagen , Colágeno/metabolismo , Progresión de la Enfermedad , Elastina/metabolismo , Femenino , Humanos , Masculino , Microvasos/diagnóstico por imagen , Microvasos/metabolismo , Países Bajos , Neuroimagen , Escalas de Valoración Psiquiátrica , Estudios Retrospectivos
13.
Blood ; 129(18): 2462-2463, 2017 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-28473412
14.
Acta Neuropathol Commun ; 12(1): 43, 2024 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-38500201

RESUMEN

Intracerebral aneurysms (IAs) are pathological dilatations of cerebral arteries whose rupture leads to subarachnoid hemorrhage, a significant cause of disability and death. Inflammation is recognized as a critical contributor to the formation, growth, and rupture of IAs; however, its precise actors have not yet been fully elucidated. Here, we report CNS-associated macrophages (CAMs), also known as border-associated macrophages, as one of the key players in IA pathogenesis, acting as critical mediators of inflammatory processes related to IA ruptures. Using a new mouse model of middle cerebral artery (MCA) aneurysms we show that CAMs accumulate in the IA walls. This finding was confirmed in a human MCA aneurysm obtained after surgical clipping, together with other pathological characteristics found in the experimental model including morphological changes and inflammatory cell infiltration. In addition, in vivo longitudinal molecular MRI studies revealed vascular inflammation strongly associated with the aneurysm area, i.e., high expression of VCAM-1 and P-selectin adhesion molecules, which precedes and predicts the bleeding extent in the case of IA rupture. Specific CAM depletion by intracerebroventricular injection of clodronate liposomes prior to IA induction reduced IA formation and rupture rate. Moreover, the absence of CAMs ameliorated the outcome severity of IA ruptures resulting in smaller hemorrhages, accompanied by reduced neutrophil infiltration. Our data shed light on the unexplored role of CAMs as main actors orchestrating the progression of IAs towards a rupture-prone state.


Asunto(s)
Aneurisma Roto , Aneurisma Intracraneal , Ratones , Animales , Humanos , Aneurisma Intracraneal/etiología , Aneurisma Intracraneal/metabolismo , Aneurisma Intracraneal/patología , Inflamación/patología , Sistema Nervioso Central/metabolismo , Factores de Riesgo , Macrófagos/metabolismo , Aneurisma Roto/complicaciones , Aneurisma Roto/metabolismo , Aneurisma Roto/patología
15.
Methods Mol Biol ; 2561: 87-101, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36399266

RESUMEN

Cerebrovascular dysfunction is a hallmark of Alzheimer's disease (AD) that is linked to cognitive decline. However, blood-brain barrier (BBB) disruption in AD is focal and requires sensitive methods to detect extravasated blood proteins and vasculature in large brain volumes. Fibrinogen, a blood coagulation factor, is deposited in AD brains at sites of BBB disruption and cerebrovascular damage. This chapter presents the methodology of fibrinogen immunolabeling-enabled three-dimensional (3D) imaging of solvent-cleared organs (iDISCO) which, when combined with immunolabeling of amyloid ß (Aß) and vasculature, enables sensitive detection of focal BBB vascular abnormalities, and reveals the spatial distribution of Aß plaques and fibrin deposits, in large tissue volumes from cleared human brains. Overall, fibrinogen iDISCO enables the investigation of neurovascular and neuroimmune mechanisms driving neurodegeneration in disease.


Asunto(s)
Enfermedad de Alzheimer , Humanos , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Fibrinógeno/metabolismo , Imagenología Tridimensional , Placa Amiloide
16.
Acta Neuropathol ; 122(3): 293-311, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21688176

RESUMEN

Cerebrovascular lesions related to congophilic amyloid angiopathy (CAA) often accompany deposition of ß-amyloid (Aß) in Alzheimer's disease (AD), leading to disturbed cerebral blood flow and cognitive dysfunction, posing the question how cerebrovascular pathology contributes to the pathology of AD. To address this question, we characterised the morphology, biochemistry and functionality of brain blood vessels in transgenic arctic ß-amyloid (arcAß) mice expressing human amyloid precursor protein (APP) with both the familial AD-causing Swedish and Arctic mutations; these mice are characterised by strong CAA pathology. Mice were analysed at early, mid and late-stage pathology. Expression of the glucose transporter GLUT1 at the blood-brain barrier (BBB) was significantly decreased and paralleled by impaired in vivo blood-to-brain glucose transport and reduced cerebral lactate release during neuronal activation from mid-stage pathology onwards. Reductions in astrocytic GLUT1 and lactate transporters, as well as retraction of astrocyte endfeet and swelling consistent with neurovascular uncoupling, preceded wide-spread ß-amyloid plaque pathology. We show that CAA at later disease stages is accompanied by severe morphological alterations of brain blood vessels including stenoses, BBB leakages and the loss of vascular smooth muscle cells (SMCs). Together, our data establish that cerebrovascular and astrocytic pathology are paralleled by impaired cerebral metabolism in arcAß mice, and that astrocyte alterations occur already at premature stages of pathology, suggesting that astrocyte dysfunction can contribute to early behavioural and cognitive impairments seen in these mice.


Asunto(s)
Astrocitos/patología , Angiopatía Amiloide Cerebral/complicaciones , Trastornos Cerebrovasculares/etiología , Trastornos Cerebrovasculares/patología , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/ultraestructura , Membrana Basal/metabolismo , Membrana Basal/patología , Barrera Hematoencefálica/patología , Barrera Hematoencefálica/fisiopatología , Encéfalo/patología , Técnicas de Cultivo de Célula , Angiopatía Amiloide Cerebral/genética , Angiopatía Amiloide Cerebral/patología , Arterias Cerebrales/metabolismo , Arterias Cerebrales/patología , Arterias Cerebrales/ultraestructura , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Distroglicanos/metabolismo , Endotelio/metabolismo , Endotelio/patología , Regulación de la Expresión Génica/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Glucosa/metabolismo , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 1/metabolismo , Hemorragia/etiología , Hemorragia/metabolismo , Hemorragia/patología , Humanos , Lactasa/metabolismo , Laminina/metabolismo , Ratones , Ratones Transgénicos , Microdiálisis/métodos , Microscopía Electrónica de Rastreo/métodos , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Músculo Liso/metabolismo , Músculo Liso/patología , Músculo Liso/ultraestructura , Placa Amiloide/metabolismo , Placa Amiloide/patología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Simportadores/genética , Simportadores/metabolismo
17.
STAR Protoc ; 2(3): 100638, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34258598

RESUMEN

Deposition of the blood coagulation factor fibrinogen in the central nervous system is a hallmark of neurological diseases with blood-brain barrier disruption. We describe in vivo two-photon imaging of microglial responses and neuronal spine elimination to either intracortical microinjection of fibrinogen in healthy mice or to endogenously labeled fibrinogen deposits in Alzheimer's disease mice. This protocol allows the longitudinal study of glial and neuronal responses to blood proteins and can be used to test drug efficacy at the neurovascular interface. For complete details on the use and execution of this protocol, please refer to Davalos et al. (2012), Ryu et al. (2018), and Merlini et al. (2019).


Asunto(s)
Encéfalo/metabolismo , Espinas Dendríticas/metabolismo , Fibrinógeno/metabolismo , Microglía/metabolismo , Microscopía/métodos , Enfermedad de Alzheimer/metabolismo , Animales , Barrera Hematoencefálica , Modelos Animales de Enfermedad , Colorantes Fluorescentes/química , Ratones , Fotones
18.
Nat Neurosci ; 24(1): 19-23, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33318667

RESUMEN

Microglial surveillance is a key feature of brain physiology and disease. Here, we found that Gi-dependent microglial dynamics prevent neuronal network hyperexcitability. By generating MgPTX mice to genetically inhibit Gi in microglia, we show that sustained reduction of microglia brain surveillance and directed process motility induced spontaneous seizures and increased hypersynchrony after physiologically evoked neuronal activity in awake adult mice. Thus, Gi-dependent microglia dynamics may prevent hyperexcitability in neurological diseases.


Asunto(s)
Quinasa 1 del Receptor Acoplado a Proteína-G/fisiología , Microglía/fisiología , Red Nerviosa/fisiología , Animales , Señalización del Calcio , Movimiento Celular , Convulsivantes , Electroencefalografía , Vigilancia Inmunológica , Ratones , Microglía/enzimología , Microglía/ultraestructura , Enfermedades del Sistema Nervioso/fisiopatología , Fenómenos Fisiológicos del Sistema Nervioso , Pilocarpina , Convulsiones/fisiopatología , Transducción de Señal , Proteínas de Unión al GTP rho/metabolismo
19.
Neuron ; 101(6): 1099-1108.e6, 2019 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-30737131

RESUMEN

Cerebrovascular alterations are a key feature of Alzheimer's disease (AD) pathogenesis. However, whether vascular damage contributes to synaptic dysfunction and how it synergizes with amyloid pathology to cause neuroinflammation and cognitive decline remain poorly understood. Here, we show that the blood protein fibrinogen induces spine elimination and promotes cognitive deficits mediated by CD11b-CD18 microglia activation. 3D molecular labeling in cleared mouse and human AD brains combined with repetitive in vivo two-photon imaging showed focal fibrinogen deposits associated with loss of dendritic spines independent of amyloid plaques. Fibrinogen-induced spine elimination was prevented by inhibiting reactive oxygen species (ROS) generation or genetic ablation of CD11b. Genetic elimination of the fibrinogen binding motif to CD11b reduced neuroinflammation, synaptic deficits, and cognitive decline in the 5XFAD mouse model of AD. Thus, fibrinogen-induced spine elimination and cognitive decline via CD11b link cerebrovascular damage with immune-mediated neurodegeneration and may have important implications in AD and related conditions.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Disfunción Cognitiva/metabolismo , Espinas Dendríticas/metabolismo , Fibrinógeno/metabolismo , Microglía/metabolismo , Placa Amiloide/metabolismo , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Encéfalo/fisiología , Encéfalo/fisiopatología , Antígeno CD11b/metabolismo , Antígenos CD18/metabolismo , Disfunción Cognitiva/patología , Disfunción Cognitiva/fisiopatología , Espinas Dendríticas/patología , Modelos Animales de Enfermedad , Humanos , Imagenología Tridimensional , Ratones , Placa Amiloide/patología , Especies Reactivas de Oxígeno/metabolismo
20.
J Cereb Blood Flow Metab ; 39(11): 2233-2245, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30073881

RESUMEN

Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) is implicated in cardiovascular disease by modulating apoptosis and oxidative stress. We hypothesized that LOX-1 may be involved in pathophysiology of stroke by mediating ischaemia/reperfusion (I/R)-dependent cell death. Transient middle cerebral artery occlusion (tMCAO) was performed in wild-type (WT) mice, endothelial-specific LOX-1 transgenic mice (eLOX-1TG) and WT animals treated with LOX-1 silencing RNA (siRNA). In WT mice exposed to tMCAO, LOX-1 expression and function were increased in the MCA. Compared to WT animals, eLOX-1TG mice displayed increased stroke volumes and worsened outcome after I/R. Conversely, LOX-1-silencing decreased both stroke volume and neurological impairment. Similarly, in HBMVECs, hypoxia/reoxygenation increased LOX-1 expression, while LOX-1 overexpressing cells showed increased death following hypoxia reoxygenation. Increased caspase-3 activation was observed following LOX-1 overexpression both in vivo and in vitro, thus representing a likely mediator. Finally, monocytes from ischaemic stroke patients exhibited increased LOX-1 expression which also correlated with disease severity. Our data unequivocally demonstrate a key role for LOX-1 in determining outcome following I/R brain damage. Our findings could be corroborated in human brain endothelial cells and monocytes from patients, underscoring their translational relevance and suggesting siRNA-mediated LOX-1 knockdown as a novel therapeutic strategy for stroke patients.


Asunto(s)
Encéfalo/patología , Daño por Reperfusión/etiología , Receptores Depuradores de Clase E/fisiología , Accidente Cerebrovascular/etiología , Animales , Apoptosis , Lesiones Encefálicas/etiología , Células Cultivadas , Células Endoteliales/metabolismo , Humanos , Infarto de la Arteria Cerebral Media , Ratones , Ratones Transgénicos , Monocitos/metabolismo , Estrés Oxidativo , ARN Interferente Pequeño/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA