Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.222
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Arterioscler Thromb Vasc Biol ; 44(8): 1737-1747, 2024 08.
Artículo en Inglés | MEDLINE | ID: mdl-38868939

RESUMEN

Dysfunctional endothelium is increasingly recognized as a mechanistic link between cardiovascular risk factors and dementia, including Alzheimer disease. BACE1 (ß-site amyloid-ß precursor protein-cleaving enzyme 1) is responsible for ß-processing of APP (amyloid-ß precursor protein), the first step in the production of Aß (amyloid-ß) peptides, major culprits in the pathogenesis of Alzheimer disease. Under pathological conditions, excessive activation of BACE1 exerts detrimental effects on endothelial function by Aß-dependent and Aß-independent mechanisms. High local concentration of Aß in the brain blood vessels is responsible for the loss of key vascular protective functions of endothelial cells. More recent studies recognized significant contribution of Aß-independent proteolytic activity of endothelial BACE1 to the pathogenesis of endothelial dysfunction. This review critically evaluates existing evidence supporting the concept that excessive activation of BACE1 expressed in the cerebrovascular endothelium impairs key homeostatic functions of the brain blood vessels. This concept has important therapeutic implications. Indeed, improved understanding of the mechanisms of endothelial dysfunction may help in efforts to develop new approaches to the protection and preservation of healthy cerebrovascular function.


Asunto(s)
Enfermedad de Alzheimer , Secretasas de la Proteína Precursora del Amiloide , Ácido Aspártico Endopeptidasas , Endotelio Vascular , Humanos , Ácido Aspártico Endopeptidasas/metabolismo , Ácido Aspártico Endopeptidasas/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Endotelio Vascular/fisiopatología , Endotelio Vascular/metabolismo , Enfermedad de Alzheimer/fisiopatología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/metabolismo , Circulación Cerebrovascular , Células Endoteliales/metabolismo , Células Endoteliales/enzimología , Células Endoteliales/patología , Encéfalo/metabolismo , Encéfalo/fisiopatología , Encéfalo/irrigación sanguínea , Encéfalo/enzimología , Trastornos Cerebrovasculares/fisiopatología , Trastornos Cerebrovasculares/metabolismo , Trastornos Cerebrovasculares/enzimología , Trastornos Cerebrovasculares/etiología
2.
Brain ; 147(9): 2946-2965, 2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-38748848

RESUMEN

Alzheimer's disease, the most prevalent form of dementia, imposes a substantial societal burden. The persistent inadequacy of disease-modifying drugs targeting amyloid plaques and neurofibrillary tangles suggests the contribution of alternative pathogenic mechanisms. A frequently overlooked aspect is cerebrovascular dysfunction, which may manifest early in the progression of Alzheimer's disease pathology. Mounting evidence underscores the pivotal role of the apolipoprotein E gene, particularly the apolipoprotein ε4 allele as the strongest genetic risk factor for late-onset Alzheimer's disease, in the cerebrovascular pathology associated with Alzheimer's disease. In this review, we examine the evidence elucidating the cerebrovascular impact of both central and peripheral apolipoprotein E on the pathogenesis of Alzheimer's disease. We present a novel three-hit hypothesis, outlining potential mechanisms that shed light on the intricate relationship among different pathogenic events. Finally, we discuss prospective therapeutics targeting the cerebrovascular pathology associated with apolipoprotein E and explore their implications for future research endeavours.


Asunto(s)
Enfermedad de Alzheimer , Apolipoproteínas E , Humanos , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Animales , Trastornos Cerebrovasculares/genética , Trastornos Cerebrovasculares/metabolismo
3.
Glia ; 72(2): 375-395, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-37909242

RESUMEN

White matter abnormalities, related to poor cerebral perfusion, are a core feature of small vessel cerebrovascular disease, and critical determinants of vascular cognitive impairment and dementia. Despite this importance there is a lack of treatment options. Proliferation of microglia producing an expanded, reactive population and associated neuroinflammatory alterations have been implicated in the onset and progression of cerebrovascular white matter disease, in patients and in animal models, suggesting that targeting microglial proliferation may exert protection. Colony-stimulating factor-1 receptor (CSF1R) is a key regulator of microglial proliferation. We found that the expression of CSF1R/Csf1r and other markers indicative of increased microglial abundance are significantly elevated in damaged white matter in human cerebrovascular disease and in a clinically relevant mouse model of chronic cerebral hypoperfusion and vascular cognitive impairment. Using the mouse model, we investigated long-term pharmacological CSF1R inhibition, via GW2580, and demonstrated that the expansion of microglial numbers in chronic hypoperfused white matter is prevented. Transcriptomic analysis of hypoperfused white matter tissue showed enrichment of microglial and inflammatory gene sets, including phagocytic genes that were the predominant expression modules modified by CSF1R inhibition. Further, CSF1R inhibition attenuated hypoperfusion-induced white matter pathology and rescued spatial learning impairments and to a lesser extent cognitive flexibility. Overall, this work suggests that inhibition of CSF1R and microglial proliferation mediates protection against chronic cerebrovascular white matter pathology and cognitive deficits. Our study nominates CSF1R as a target for the treatment of vascular cognitive disorders with broader implications for treatment of other chronic white matter diseases.


Asunto(s)
Trastornos Cerebrovasculares , Trastornos del Conocimiento , Disfunción Cognitiva , Leucoencefalopatías , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos , Sustancia Blanca , Animales , Ratones , Trastornos Cerebrovasculares/metabolismo , Trastornos Cerebrovasculares/patología , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/patología , Disfunción Cognitiva/metabolismo , Modelos Animales de Enfermedad , Leucoencefalopatías/genética , Leucoencefalopatías/metabolismo , Ratones Endogámicos C57BL , Microglía/metabolismo , Receptores del Factor Estimulante de Colonias/metabolismo , Sustancia Blanca/patología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo
4.
Clin Sci (Lond) ; 138(20): 1305-1323, 2024 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-39391895

RESUMEN

Symmetric dimethylarginine (SDMA) is a methylated derivative of arginine, generated by all cells as a by-product of cellular metabolism and eliminated via the kidney. For many years SDMA has been considered inert and of little biological significance. However, a growing body of evidence now suggests this view is outdated and that circulating SDMA levels may, in fact, be intricately linked to endothelial dysfunction and vascular risk. In this review, we specifically examine SDMA within the context of cerebrovascular disease, with a particular focus on ischaemic stroke. We first discuss pre-clinical evidence supporting the notion that SDMA has effects on nitric oxide signalling, inflammation, oxidative stress, and HDL function. We then appraise the most recent clinical studies that explore the relationship between circulating SDMA and cerebrovascular risk factors, such as chronic kidney disease, hypertension, atrial fibrillation, and atherosclerosis, exploring whether any associations may arise due to the existence of shared risk factors. Finally, we consider the evidence that elevated circulating SDMA is linked to poor outcomes following ischaemic and haemorrhagic stroke. We draw upon pre-clinical insights into SDMA function to speculate how SDMA may not only be a marker of cerebrovascular disease but could also directly influence cerebrovascular pathology, and we highlight the pressing need for more mechanistic pre-clinical studies alongside adequately powered, longitudinal clinical studies to fully evaluate SDMA as a marker/mediator of disease.


Asunto(s)
Arginina , Biomarcadores , Trastornos Cerebrovasculares , Humanos , Biomarcadores/sangre , Trastornos Cerebrovasculares/sangre , Trastornos Cerebrovasculares/metabolismo , Arginina/análogos & derivados , Arginina/sangre , Arginina/metabolismo , Animales , Factores de Riesgo , Estrés Oxidativo , Accidente Cerebrovascular Isquémico/sangre , Accidente Cerebrovascular Isquémico/metabolismo
5.
Nitric Oxide ; 152: 48-57, 2024 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-39299647

RESUMEN

Nitric oxide (NO) plays a crucial role as a messenger or effector in the body, yet it presents a dual impact on cardio-cerebrovascular health. Under normal physiological conditions, NO exhibits vasodilatory effects, regulates blood pressure, inhibits platelet aggregation, and offers neuroprotective actions. However, in pathological situations, excessive NO production contributes to or worsens inflammation within the body. Moreover, NO may combine with reactive oxygen species (ROS), generating harmful substances that intensify physical harm. This paper succinctly reviews pertinent literature to clarify the in vivo and in vitro origins of NO, its regulatory function in the cardio-cerebrovascular system, and the advantages and disadvantages associated with NO donor drugs, NO delivery systems, and vascular stent materials for treating cardio-cerebrovascular disease. The findings provide a theoretical foundation for the application of NO in cardio-cerebrovascular diseases.


Asunto(s)
Óxido Nítrico , Óxido Nítrico/metabolismo , Humanos , Animales , Trastornos Cerebrovasculares/metabolismo , Trastornos Cerebrovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/metabolismo , Donantes de Óxido Nítrico/farmacología , Donantes de Óxido Nítrico/química , Sistema Cardiovascular/metabolismo , Sistema Cardiovascular/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo
6.
Biol Pharm Bull ; 47(1): 104-111, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38171771

RESUMEN

White matter lesions induced by chronic cerebral hypoperfusion can cause vascular dementia; however, no appropriate treatments are currently available for these diseases. In this study, we investigated lipid peroxidation, which has recently been pointed out to be associated with cerebrovascular disease and vascular dementia, as a therapeutic target for chronic cerebral hypoperfusion. We used ethoxyquin, a lipid-soluble antioxidant, in a neuronal cell line and mouse model of the disease. The cytoprotective effect of ethoxyquin on glutamate-stimulated HT-22 cells, a mouse hippocampal cell line, was comparable to that of a ferroptosis inhibitor. In addition, the administration of ethoxyquin to bilateral common carotid artery stenosis model mice suppressed white matter lesions, blood-brain barrier disruption, and glial cell activation. Taken together, we propose that the inhibition of lipid peroxidation may be a useful therapeutic approach for chronic cerebrovascular disease and the resulting white matter lesions.


Asunto(s)
Isquemia Encefálica , Estenosis Carotídea , Trastornos Cerebrovasculares , Demencia Vascular , Sustancia Blanca , Animales , Ratones , Demencia Vascular/complicaciones , Etoxiquina/metabolismo , Etoxiquina/farmacología , Etoxiquina/uso terapéutico , Sustancia Blanca/metabolismo , Sustancia Blanca/patología , Isquemia Encefálica/patología , Trastornos Cerebrovasculares/tratamiento farmacológico , Trastornos Cerebrovasculares/complicaciones , Trastornos Cerebrovasculares/metabolismo , Modelos Animales de Enfermedad , Estenosis Carotídea/complicaciones , Estenosis Carotídea/metabolismo , Estenosis Carotídea/patología , Ratones Endogámicos C57BL
7.
Adv Exp Med Biol ; 1463: 195-200, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39400823

RESUMEN

Endothelial dysfunction featuring insufficient endothelial nitric oxide synthase (eNOS) and accompanying nitric oxide (NO) deficiency is implicated in the pathogenesis of cardiovascular diseases. Restoring endothelial NO represents a promising approach to treating cerebrovascular diseases, including stroke. Low-power near-infrared (NIR) light shows diverse beneficial effects, broadly defined as photobiomodulation (PBM). The literature reports that PBM increases bioavailable NO. These lines of evidence indicate that PBM could be used to treat cerebrovascular diseases. Recent investigations revealed that PBM improved stroke outcomes in animal models via augmenting NO signalling and other pathways. However, clinical trials of PBM using NIR light in the NIR-I window (630-900 nm) have yet to demonstrate the beneficial effect of PBM on ischaemic stroke. Since NIR light in the NIR-II window (1000-1700 nm) with the largest penetration depth into tissues compared to NIR I has also been reported to augment NO bioavailability and cerebral blood flow ameliorating stroke injury, PBM using NIR-II light may be suitable for therapeutic use. This new non-pharmacological modality using a physical parameter of NIR-II laser could provide a new avenue for therapeutic strategies for cerebrovascular diseases. Since impaired NO production has been associated with neurological abnormalities, this novel therapeutic approach could be broadly explored to treat various disease conditions such as traumatic brain injury, stroke, and Alzheimer's disease. This review summarises recent findings on PBM in treating stroke and discusses its potential to treat other neurological diseases.


Asunto(s)
Terapia por Luz de Baja Intensidad , Óxido Nítrico , Transducción de Señal , Óxido Nítrico/metabolismo , Humanos , Terapia por Luz de Baja Intensidad/métodos , Animales , Transducción de Señal/efectos de la radiación , Trastornos Cerebrovasculares/radioterapia , Trastornos Cerebrovasculares/metabolismo , Trastornos Cerebrovasculares/fisiopatología , Rayos Infrarrojos/uso terapéutico , Óxido Nítrico Sintasa de Tipo III/metabolismo , Circulación Cerebrovascular/efectos de la radiación
8.
Molecules ; 29(9)2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38731591

RESUMEN

Angelica sinensis (Oliv.) Diels (A. sinensis) is a medicinal and edible values substance, which could promote blood circulation and enrich blood. It possesses rich chemical components and nutrients, which have significant therapeutic effects on cardiovascular and cerebrovascular diseases. It is commonly used for the prevention and treatment of cardiovascular and cerebrovascular diseases in the elderly, especially in improving ischemic damage to the heart and brain, protecting vascular cells, and regulating inflammatory reactions. This article reviews the main pharmacological effects and clinical research of A. sinensis on cardiovascular and cerebrovascular diseases in recent years, explores the effect of its chemical components on cardiovascular and cerebrovascular diseases by regulating the expression of functional proteins and inhibiting inflammation, anti-apoptosis, and antioxidant mechanisms. It provides a reference for further research on A. sinensis and the development of related drugs. It provides a new reference direction for the in-depth research and application of A. sinensis in the prevention, improvement, and treatment of cardiovascular and cerebrovascular diseases.


Asunto(s)
Angelica sinensis , Enfermedades Cardiovasculares , Trastornos Cerebrovasculares , Humanos , Angelica sinensis/química , Trastornos Cerebrovasculares/tratamiento farmacológico , Trastornos Cerebrovasculares/metabolismo , Enfermedades Cardiovasculares/tratamiento farmacológico , Animales , Antioxidantes/farmacología , Antioxidantes/química , Extractos Vegetales/farmacología , Extractos Vegetales/química
9.
FASEB J ; 36(1): e22075, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34919285

RESUMEN

Long non-coding RNAs (lncRNAs) regulate neurological damage in cerebral ischemia-reperfusion injury (CIRI). This study aimed to investigate the biological roles of lncRNA CEBPA-AS1 in CIRI. Middle cerebral artery occlusion and ischemia-reperfusion injury (MCAO/IR) rat model and oxygen-glucose deprivation and reoxygenation (OGD/R) cell lines were generated; the expression of CEBPA-AS1 was evaluated by qRT-PCR. The effects of CEBPA-AS1 on cell apoptosis and nerve damage were examined. The downstream microRNA (miRNA) and mRNA of CEBPA-AS1 were predicted and verified. We found that overexpression of CEBPA-AS1 could attenuate MCAO/IR-induced nerve damage and neuronal apoptosis in the rat model. Knockdown of CEBPA-AS1 aggravated cell apoptosis and enhanced the production of LDH and MDA in the OGD/R cells. Upon examining the molecular mechanisms, we found that CEBPA-AS1 stimulated APPL1 expression by combining with miR-340-5p, thereby regulating the APPL1/LKB1/AMPK pathway. In the rescue experiments, CEBPA-AS1 overexpression was found to attenuate OGD/R-induced cell apoptosis and MCAO/IR induced nerve damage, while miR-340-5p reversed these effects of CEBPA-AS1. In conclusion, CEBPA-AS1 could decrease CIRI by sponging miR-340-5, regulating the APPL1/LKB1/AMPK pathway.


Asunto(s)
Quinasas de la Proteína-Quinasa Activada por el AMP/biosíntesis , Proteínas Quinasas Activadas por AMP/biosíntesis , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Trastornos Cerebrovasculares/metabolismo , MicroARNs/biosíntesis , Proteínas del Tejido Nervioso/biosíntesis , ARN Largo no Codificante/biosíntesis , Daño por Reperfusión/metabolismo , Transducción de Señal , Quinasas de la Proteína-Quinasa Activada por el AMP/genética , Proteínas Quinasas Activadas por AMP/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Apoptosis , Trastornos Cerebrovasculares/genética , Trastornos Cerebrovasculares/patología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , MicroARNs/genética , Proteínas del Tejido Nervioso/genética , ARN Largo no Codificante/genética , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/genética , Daño por Reperfusión/patología
10.
Int J Mol Sci ; 24(22)2023 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-38003477

RESUMEN

The blood-brain barrier (BBB) is a unique and selective feature of the central nervous system's vasculature. BBB dysfunction has been observed as an early sign of Alzheimer's Disease (AD) before the onset of dementia or neurodegeneration. The intricate relationship between the BBB and the pathogenesis of AD, especially in the context of neurovascular coupling and the overlap of pathophysiology in neurodegenerative and cerebrovascular diseases, underscores the urgency to understand the BBB's role more deeply. Preserving or restoring the BBB function emerges as a potentially promising strategy for mitigating the progression and severity of AD. Molecular and genetic changes, such as the isoform ε4 of apolipoprotein E (ApoEε4), a significant genetic risk factor and a promoter of the BBB dysfunction, have been shown to mediate the BBB disruption. Additionally, receptors and transporters like the low-density lipoprotein receptor-related protein 1 (LRP1), P-glycoprotein (P-gp), and the receptor for advanced glycation end products (RAGEs) have been implicated in AD's pathogenesis. In this comprehensive review, we endeavor to shed light on the intricate pathogenic and therapeutic connections between AD and the BBB. We also delve into the latest developments and pioneering strategies targeting the BBB for therapeutic interventions, addressing its potential as a barrier and a carrier. By providing an integrative perspective, we anticipate paving the way for future research and treatments focused on exploiting the BBB's role in AD pathogenesis and therapy.


Asunto(s)
Enfermedad de Alzheimer , Trastornos Cerebrovasculares , Humanos , Enfermedad de Alzheimer/metabolismo , Barrera Hematoencefálica/metabolismo , Péptidos beta-Amiloides/metabolismo , Transporte Biológico/fisiología , Trastornos Cerebrovasculares/metabolismo
11.
Physiol Rev ; 95(3): 953-93, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26133935

RESUMEN

Spreading depression (SD) is a transient wave of near-complete neuronal and glial depolarization associated with massive transmembrane ionic and water shifts. It is evolutionarily conserved in the central nervous systems of a wide variety of species from locust to human. The depolarization spreads slowly at a rate of only millimeters per minute by way of grey matter contiguity, irrespective of functional or vascular divisions, and lasts up to a minute in otherwise normal tissue. As such, SD is a radically different breed of electrophysiological activity compared with everyday neural activity, such as action potentials and synaptic transmission. Seventy years after its discovery by Leão, the mechanisms of SD and its profound metabolic and hemodynamic effects are still debated. What we did learn of consequence, however, is that SD plays a central role in the pathophysiology of a number of diseases including migraine, ischemic stroke, intracranial hemorrhage, and traumatic brain injury. An intriguing overlap among them is that they are all neurovascular disorders. Therefore, the interplay between neurons and vascular elements is critical for our understanding of the impact of this homeostatic breakdown in patients. The challenges of translating experimental data into human pathophysiology notwithstanding, this review provides a detailed account of bidirectional interactions between brain parenchyma and the cerebral vasculature during SD and puts this in the context of neurovascular diseases.


Asunto(s)
Arterias Cerebrales/fisiopatología , Circulación Cerebrovascular , Trastornos Cerebrovasculares/fisiopatología , Depresión de Propagación Cortical , Potenciales de la Membrana , Vías Nerviosas/fisiopatología , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/fisiopatología , Arterias Cerebrales/metabolismo , Trastornos Cerebrovasculares/metabolismo , Metabolismo Energético , Hemodinámica , Humanos , Vías Nerviosas/metabolismo , Transducción de Señal , Factores de Tiempo
12.
Anal Biochem ; 636: 114387, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34537182

RESUMEN

Non-alcoholic fatty liver disease (NAFLD) is a multisystem disease, which has been classified as an emerging epidemic not only confined to liver-related morbidity and mortality. It is also becoming apparent that NAFLD is associated with moderate cerebral dysfunction and cognitive decline. A possible link between NAFLD and Alzheimer's disease (AD) has only recently been proposed due to the multiple shared genes and pathological mechanisms contributing to the development of these conditions. Although AD is a progressive neurodegenerative disease, the exact pathophysiological mechanism remains ambiguous and similarly to NAFLD, currently available pharmacological therapies have mostly failed in clinical trials. In addition to the usual suspects (inflammation, oxidative stress, blood-brain barrier alterations and ageing) that could contribute to the NAFLD-induced development and progression of AD, changes in the vasculature, cerebral perfusion and waste clearance could be the missing link between these two diseases. Here, we review the most recent literature linking NAFLD and AD, focusing on cerebrovascular alterations and the brain's clearance system as risk factors involved in the development and progression of AD, with the aim of promoting further research using neuroimaging techniques and new mechanism-based therapeutic interventions.


Asunto(s)
Envejecimiento/metabolismo , Enfermedad de Alzheimer , Trastornos Cerebrovasculares , Enfermedad del Hígado Graso no Alcohólico , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/fisiopatología , Trastornos Cerebrovasculares/etiología , Trastornos Cerebrovasculares/metabolismo , Trastornos Cerebrovasculares/fisiopatología , Humanos , Inflamación/metabolismo , Inflamación/fisiopatología , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/fisiopatología , Estrés Oxidativo , Factores de Riesgo
13.
J Cell Mol Med ; 25(12): 5341-5350, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33942488

RESUMEN

Sestrin2 (SESN2) is a conserved stress-inducible protein (also known as hypoxia-inducible gene 95 (HI95)) that is induced under hypoxic conditions. SESN2 represses the production of reactive oxygen species (ROS) and provides cytoprotection against various noxious stimuli, including hypoxia, oxidative stress, endoplasmic reticulum (ER) stress and DNA damage. In recent years, the determination of the regulation and signalling mechanisms of SESN2 has increased our understanding of its role in the hypoxic response. SESN2 has well-documented roles in hypoxia-related diseases, making it a potential target for diagnosis and treatment. This review discusses the regulatory mechanisms of SESN2 and highlights the significance of SESN2 as a biomarker and therapeutic target in hypoxia-related diseases, such as cancer, respiratory-related diseases, cardiovascular diseases and cerebrovascular diseases.


Asunto(s)
Enfermedades Cardiovasculares/patología , Trastornos Cerebrovasculares/patología , Hipoxia/fisiopatología , Neoplasias/patología , Proteínas Nucleares/metabolismo , Peroxidasas/metabolismo , Enfermedades Respiratorias/patología , Animales , Enfermedades Cardiovasculares/metabolismo , Trastornos Cerebrovasculares/metabolismo , Estrés del Retículo Endoplásmico , Humanos , Neoplasias/metabolismo , Proteínas Nucleares/genética , Estrés Oxidativo , Peroxidasas/genética , Especies Reactivas de Oxígeno , Enfermedades Respiratorias/metabolismo
14.
Stroke ; 52(7): 2404-2413, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34107734

RESUMEN

Background and Purpose: Hypertension is a leading risk factor for cerebrovascular disease and loss of brain health. While the brain renin-angiotensin system (RAS) contributes to hypertension, its potential impact on the local vasculature is unclear. We tested the hypothesis that activation of the brain RAS would alter the local vasculature using a modified deoxycorticosterone acetate (DOCA) model. Methods: C57BL/6 mice treated with DOCA (50 mg SQ; or shams) were given tap H2O and H2O with 0.9% NaCl for 1 to 3 weeks. Results: In isolated cerebral arteries and parenchymal arterioles from DOCA-treated male mice, endothelium- and nitric oxide-dependent dilation was progressively impaired, while mesenteric arteries were unaffected. In contrast, cerebral endothelial function was not significantly affected in female mice treated with DOCA. In males, mRNA expression of renal Ren1 was markedly reduced while RAS components (eg, Agt and Ace) were increased in both brain and cerebral arteries with central RAS activation. In NZ44 reporter mice expressing GFP (green fluorescent protein) driven by the angiotensin II type 1A receptor (Agtr1a) promoter, DOCA increased GFP expression ≈3-fold in cerebral arteries. Impaired endothelial responses were restored to normal by losartan, an AT1R (angiotensin II type 1 receptor) antagonist. Last, DOCA treatment produced inward remodeling of parenchymal arterioles. Conclusions: These findings suggest activation of the central and cerebrovascular RAS impairs endothelial (nitric oxide dependent) signaling in brain through expression and activation of AT1R and sex-dependent effects. The central RAS may be a key contributor to vascular dysfunction in brain in a preclinical (low renin) model of hypertension. Because the brain RAS is also activated during aging and other diseases, a common mechanism may promote loss of endothelial and brain health despite diverse cause.


Asunto(s)
Trastornos Cerebrovasculares/metabolismo , Endotelio Vascular/metabolismo , Hipertensión/metabolismo , Óxido Nítrico Sintasa de Tipo III/biosíntesis , Receptor de Angiotensina Tipo 1/biosíntesis , Sistema Renina-Angiotensina/fisiología , Animales , Trastornos Cerebrovasculares/inducido químicamente , Trastornos Cerebrovasculares/genética , Acetato de Desoxicorticosterona/toxicidad , Femenino , Hipertensión/inducido químicamente , Hipertensión/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Óxido Nítrico Sintasa de Tipo III/genética , Receptor de Angiotensina Tipo 1/genética , Sistema Renina-Angiotensina/efectos de los fármacos
15.
Neuroimage ; 233: 117955, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33716155

RESUMEN

Cerebrovascular reactivity (CVR) reflects the capacity of the brain to meet changing physiological demands and can predict the risk of cerebrovascular diseases. CVR can be obtained by measuring the change in cerebral blood flow (CBF) during a brain stress test where CBF is altered by a vasodilator such as acetazolamide. Although the gold standard to quantify CBF is PET imaging, the procedure is invasive and inaccessible to most patients. Arterial spin labeling (ASL) is a non-invasive and quantitative MRI method to measure CBF, and a consensus guideline has been published for the clinical application of ASL. Despite single post labeling delay (PLD) pseudo-continuous ASL (PCASL) being the recommended ASL technique for CBF quantification, it is sensitive to variations to the arterial transit time (ATT) and labeling efficiency induced by the vasodilator in CVR studies. Multi-PLD ASL controls for the changes in ATT, and velocity selective ASL is in theory insensitive to both ATT and labeling efficiency. Here we investigate CVR using simultaneous 15O-water PET and ASL MRI data from 19 healthy subjects. CVR and CBF measured by the ASL techniques were compared using PET as the reference technique. The impacts of blood T1 and labeling efficiency on ASL were assessed using individual measurements of hematocrit and flow velocity data of the carotid and vertebral arteries measured using phase-contrast MRI. We found that multi-PLD PCASL is the ASL technique most consistent with PET for CVR quantification (group mean CVR of the whole brain = 42±19% and 40±18% respectively). Single-PLD ASL underestimated the CVR of the whole brain significantly by 15±10% compared with PET (p<0.01, paired t-test). Changes in ATT pre- and post-acetazolamide was the principal factor affecting ASL-based CVR quantification. Variations in labeling efficiency and blood T1 had negligible effects.


Asunto(s)
Velocidad del Flujo Sanguíneo/fisiología , Encéfalo/metabolismo , Trastornos Cerebrovasculares/metabolismo , Imagen por Resonancia Magnética/normas , Tomografía de Emisión de Positrones/normas , Marcadores de Spin , Adulto , Anciano , Encéfalo/diagnóstico por imagen , Circulación Cerebrovascular/fisiología , Trastornos Cerebrovasculares/diagnóstico por imagen , Femenino , Hematócrito/métodos , Hematócrito/normas , Humanos , Imagen por Resonancia Magnética/métodos , Masculino , Persona de Mediana Edad , Radioisótopos de Oxígeno/metabolismo , Tomografía de Emisión de Positrones/métodos , Factores de Tiempo , Agua/metabolismo
16.
J Neurochem ; 156(4): 539-552, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32683713

RESUMEN

Systemic multimorbidity is highly prevalent in the elderly and, remarkably, coexisting neuropathological markers of Alzheimer's (AD) and cerebrovascular (CVD) diseases are found at autopsy in most brains of patients clinically diagnosed as AD. Little is known on neurodegeneration peculiar to comorbidities, especially at early stages when pathogenesis may propagate at subclinical levels. We developed a novel in vitro model of comorbid CVD/AD in organotypic hippocampal cultures, by combining oxygen-glucose deprivation (OGD) and exposure to amyloid-Aß oligomers (AßOs), both applied at levels subtoxic to neurons when used in isolation. We focused on synaptic proteins and the roles of glutamate receptors, which have been implicated in many basic and clinical approaches to either CVD or AD. Subtoxic insults by OGD and AßOs synergized to reduce levels of synaptophysin (SYP) and PSD-95 without cell death, while effects of antagonists of either metabotropic or ionotropic glutamate receptors were distinct from reports in models of isolated CVD or AD. In particular, modulation of glutamate receptors differentially impacted SYP and PSD-95, and antagonists of a single receptor subtype had distinct effects when either isolated or combined. Our findings highlight the complexity of CVD/AD comorbidity, help understand variable responses to glutamate receptor antagonists in patients diagnosed with AD and may contribute to future development of therapeutics based on investigation of the pattern of progressive comorbidity.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Trastornos Cerebrovasculares/metabolismo , Hipocampo/metabolismo , Receptores de Glutamato/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Animales , Muerte Celular/fisiología , Hipoxia de la Célula/fisiología , Trastornos Cerebrovasculares/genética , Trastornos Cerebrovasculares/patología , Comorbilidad , Glucosa/deficiencia , Hipocampo/patología , Masculino , Técnicas de Cultivo de Órganos , Ratas , Receptores de Glutamato/genética
17.
J Neurochem ; 158(3): 724-736, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-32441775

RESUMEN

Cerebrovascular-related amyloidogenesis is found in over 80% of Alzheimer's disease (AD) cases, and amyloid ß (Aß) generation is increased in the peripheral macrophages during infection of Porphyromonas gingivalis (P. gingivalis), a causal bacterium for periodontitis. In this study, we focused on receptor for advanced glycation end products (RAGE), the key molecule involves in Aß influx after P. gingivalis infection to test our hypothesis that Aß transportation from periphery into the brain, known as "Aß influx," is enhanced by P. gingivalis infection. Using cultured hCMEC/D3 cell line, in comparison to uninfected cells, directly infection with P. gingivalis (multiplicity of infection, MOI = 5) significantly increased a time-dependent RAGE expression resulting in a dramatic increase in Aß influx in the hCMEC/D3 cells; the P. gingivalis-up-regulated RAGE expression was significantly decreased by NF-κB and Cathepsin B (CatB)-specific inhibitors, and the P.gingivalis-increased IκBα degradation was significantly decreased by CatB-specific inhibitor. Furthermore, the P. gingivalis-increased Aß influx was significantly reduced by RAGE-specific inhibitor. Using 15-month-old mice (C57BL/6JJmsSlc, female), in comparison to non-infection mice, systemic P. gingivalis infection for three consecutive weeks (1 × 108  CFU/mouse, every 3 days, intraperitoneally) significantly increased the RAGE expression in the CD31-positive endothelial cells and the Aß loads around the CD31-positive cells in the mice's brains. The RAGE expression in the CD31-positive cells was positively correlated with the Aß loads. These observations demonstrate that the up-regulated RAGE expression in cerebral endothelial cells mediates the Aß influx after P. gingivalis infection, and CatB plays a critical role in regulating the NF-κB/RAGE expression. Cover Image for this issue: https://doi.org/10.1111/jnc.15073.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Infecciones por Bacteroidaceae/metabolismo , Corteza Cerebral/metabolismo , Células Endoteliales/metabolismo , Fragmentos de Péptidos/metabolismo , Porphyromonas gingivalis , Receptor para Productos Finales de Glicación Avanzada/biosíntesis , Animales , Corteza Cerebral/microbiología , Circulación Cerebrovascular/fisiología , Trastornos Cerebrovasculares/metabolismo , Trastornos Cerebrovasculares/microbiología , Células Endoteliales/microbiología , Femenino , Ratones , Ratones Endogámicos C57BL , Regulación hacia Arriba/fisiología
18.
Biochem Biophys Res Commun ; 555: 89-94, 2021 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-33813281

RESUMEN

Sporadic Alzheimer's disease (sAD) is the commonest cause of age-related neurodegeneration but there are no available treatments with demonstrated disease-modifying actions. It is therefore relevant to study hitherto-unknown aspects of brain structure and function to seek new disease-related mechanisms that might be targeted by novel disease-modifying interventions. During hypothesis-generating proteomic investigations in a case-control study of sAD, we observed widespread elevations of haptoglobin and haemopexin in all six brain-regions studied, which together represent much of the brain. Measured perturbations were significant, with the posterior probability of upregulation generally >95% and haptoglobin doubling in expression levels on average across deep brain structures (hippocampus, entorhinal cortex and cingulate gyrus) as well as sensory and motor cortices, and cerebellum. Haptoglobin and haemopexin are often regarded as circulating proteins whose main functions are to bind, respectively, the strongly pro-inflammatory extracellular haemoglobin and haeme molecules that form following haemolysis, thereby promoting their clearance and suppressing damage they might otherwise cause, for example, acute kidney injury. To our knowledge, elevations in neither cerebral haptoglobin nor haemopexin have previously been linked to the pathogenesis of sAD. Post-mortem examination of these cases showed no signs of macroscopic cerebral haemorrhage. These findings demonstrate pervasive cerebral elevation of haptoglobin and haemopexin, consistent with low-level intracerebral leakage of haemoglobin and consequent haeme formation throughout sAD brain. They point to a widespread underlying microvasculopathy that facilitates erythrocyte leakage, thereby triggering elevated tissue-free haemoglobin and driving the measured elevations in haptoglobin and haemopexin.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Haptoglobinas/análisis , Hemopexina/análisis , Anciano , Barrera Hematoencefálica/fisiopatología , Encéfalo/irrigación sanguínea , Encéfalo/fisiopatología , Estudios de Casos y Controles , Trastornos Cerebrovasculares/metabolismo , Trastornos Cerebrovasculares/fisiopatología , Femenino , Humanos , Hierro/análisis , Hierro/metabolismo , Masculino
19.
Basic Res Cardiol ; 116(1): 23, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33770265

RESUMEN

Despite major advances in prevention and treatment, cardiac and cerebral atherothrombotic complications still account for substantial morbidity and mortality worldwide. In this context, inflammation is involved in the chronic process leading atherosclerotic plaque formation and its complications, as well as in the maladaptive response to acute ischemic events. For this reason, modulation of inflammation is nowadays seen as a promising therapeutic strategy to counteract the burden of cardio- and cerebrovascular disease. Being produced and recognized by both inflammatory and vascular cells, the complex network of cytokines holds key functions in the crosstalk of these two systems and orchestrates the progression of atherothrombosis. By binding to membrane receptors, these soluble mediators trigger specific intracellular signaling pathways eventually leading to the activation of transcription factors and a deep modulation of cell function. Both stimulatory and inhibitory cytokines have been described and progressively reported as markers of disease or interesting therapeutic targets in the cardiovascular field. Nevertheless, cytokine inhibition is burdened by harmful side effects that will most likely prevent its chronic use in favor of acute administrations in well-selected subjects at high risk. Here, we summarize the current state of knowledge regarding the modulatory role of cytokines on atherosclerosis, myocardial infarction, and stroke. Then, we discuss evidence from clinical trials specifically targeting cytokines and the potential implication of these advances into daily clinical practice.


Asunto(s)
Antiinflamatorios/uso terapéutico , Enfermedades Cardiovasculares/tratamiento farmacológico , Sistema Cardiovascular/efectos de los fármacos , Trastornos Cerebrovasculares/tratamiento farmacológico , Citocinas/antagonistas & inhibidores , Mediadores de Inflamación/antagonistas & inhibidores , Inflamación/tratamiento farmacológico , Animales , Antiinflamatorios/efectos adversos , Enfermedades Cardiovasculares/inmunología , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/fisiopatología , Sistema Cardiovascular/inmunología , Sistema Cardiovascular/metabolismo , Sistema Cardiovascular/fisiopatología , Trastornos Cerebrovasculares/inmunología , Trastornos Cerebrovasculares/metabolismo , Trastornos Cerebrovasculares/fisiopatología , Citocinas/metabolismo , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/fisiopatología , Mediadores de Inflamación/metabolismo , Terapia Molecular Dirigida , Transducción de Señal
20.
FASEB J ; 34(7): 9575-9593, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32472598

RESUMEN

The obesity epidemic increases the interest to elucidate impact of short-chain fatty acids on metabolism, obesity, and the brain. We investigated the effects of propionic acid (PA) and caproic acid (CA) on metabolic risk factors, liver and adipose tissue pathology, brain function, structure (by MRI), and gene expression, during obesity development in Ldlr-/- .Leiden mice. Ldlr-/- .Leiden mice received 16 weeks either a high-fat diet (HFD) to induce obesity, or chow as reference group. Next, obese HFD-fed mice were treated 12 weeks with (a) HFD + CA (CA), (b) HFD + PA (PA), or (c) a HFD-control group. PA reduced the body weight and systolic blood pressure, lowered fasting insulin levels, and reduced HFD-induced liver macrovesicular steatosis, hypertrophy, inflammation, and collagen content. PA increased the amount of glucose transporter type 1-positive cerebral blood vessels, reverted cerebral vasoreactivity, and HFD-induced effects in microstructural gray and white matter integrity of optic tract, and somatosensory and visual cortex. PA and CA also reverted HFD-induced effects in functional connectivity between visual and auditory cortex. However, PA mice were more anxious in open field, and showed reduced activity of synaptogenesis and glutamate regulators in hippocampus. Therefore, PA treatment should be used with caution even though positive metabolic, (cerebro) vascular, and brain structural and functional effects were observed.


Asunto(s)
Caproatos/farmacología , Trastornos Cerebrovasculares/prevención & control , Inflamación/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Obesidad/complicaciones , Propionatos/farmacología , Receptores de LDL/fisiología , Animales , Trastornos Cerebrovasculares/etiología , Trastornos Cerebrovasculares/metabolismo , Trastornos Cerebrovasculares/patología , Dieta con Restricción de Grasas/efectos adversos , Dieta Alta en Grasa/efectos adversos , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones , Ratones Noqueados , Ratones Obesos , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA