Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Redox Biol ; 24: 101192, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31015147

RESUMEN

Adjustment of cerebral blood flow (CBF) to neuronal activity via neurovascular coupling (NVC) has an essential role in maintenance of healthy cognitive function. In aging increased oxidative stress and cerebromicrovascular endothelial dysfunction impair NVC, contributing to cognitive decline. There is increasing evidence showing that a decrease in NAD+ availability with age plays a critical role in a range of age-related cellular impairments but its role in impaired NVC responses remains unexplored. The present study was designed to test the hypothesis that restoring NAD+ concentration may exert beneficial effects on NVC responses in aging. To test this hypothesis 24-month-old C57BL/6 mice were treated with nicotinamide mononucleotide (NMN), a key NAD+ intermediate, for 2 weeks. NVC was assessed by measuring CBF responses (laser Doppler flowmetry) evoked by contralateral whisker stimulation. We found that NVC responses were significantly impaired in aged mice. NMN supplementation rescued NVC responses by increasing endothelial NO-mediated vasodilation, which was associated with significantly improved spatial working memory and gait coordination. These findings are paralleled by the sirtuin-dependent protective effects of NMN on mitochondrial production of reactive oxygen species and mitochondrial bioenergetics in cultured cerebromicrovascular endothelial cells derived from aged animals. Thus, a decrease in NAD+ availability contributes to age-related cerebromicrovascular dysfunction, exacerbating cognitive decline. The cerebromicrovascular protective effects of NMN highlight the preventive and therapeutic potential of NAD+ intermediates as effective interventions in patients at risk for vascular cognitive impairment (VCI).


Asunto(s)
Circulación Cerebrovascular , Disfunción Cognitiva , Suplementos Dietéticos , Endotelio Vascular/metabolismo , Acoplamiento Neurovascular , Mononucleótido de Nicotinamida/administración & dosificación , Factores de Edad , Animales , Conducta Animal , Biomarcadores , Disfunción Cognitiva/tratamiento farmacológico , Humanos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Mitocondrias/metabolismo , Óxido Nítrico/metabolismo , Estrés Oxidativo/efectos de los fármacos , ARN Interferente Pequeño/genética , Especies Reactivas de Oxígeno/metabolismo
2.
J Gerontol A Biol Sci Med Sci ; 74(3): 290-298, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29893815

RESUMEN

There is strong evidence that obesity has deleterious effects on cognitive function of older adults. Previous preclinical studies demonstrate that obesity in aging is associated with a heightened state of systemic inflammation, which exacerbates blood-brain barrier disruption, promoting neuroinflammation and oxidative stress. To test the hypothesis that synergistic effects of obesity and aging on inflammatory processes exert deleterious effects on hippocampal function, young and aged C57BL/6 mice were rendered obese by chronic feeding of a high-fat diet followed by assessment of learning and memory function, measurement of hippocampal long-term potentiation (LTP), assessment of changes in hippocampal expression of genes relevant for synaptic function and determination of synaptic density. Because there is increasing evidence that altered production of lipid mediators modulate LTP, neuroinflammation and neurovascular coupling responses, the effects of obesity on hippocampal levels of relevant eicosanoid mediators were also assessed. We found that aging exacerbates obesity-induced microglia activation, which is associated with deficits in hippocampal-dependent learning and memory tests, impaired LTP, decreased synaptic density, and dysregulation of genes involved in regulation of synaptic plasticity. Obesity in aging also resulted in an altered hippocampal eicosanoid profile, including decreases in vasodilator and pro-LTP epoxy-eicosatrienoic acids (EETs). Collectively, our results taken together with previous findings suggest that obesity in aging promotes hippocampal inflammation, which in turn may contribute to synaptic dysfunction and cognitive impairment.


Asunto(s)
Envejecimiento/fisiología , Disfunción Cognitiva/etiología , Eicosanoides/metabolismo , Hipocampo/metabolismo , Plasticidad Neuronal/fisiología , Obesidad/psicología , Animales , Biomarcadores/sangre , Biomarcadores/metabolismo , Disfunción Cognitiva/fisiopatología , Modelos Animales de Enfermedad , Inflamación , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/complicaciones , Obesidad/metabolismo , Estrés Oxidativo/fisiología
3.
J Gerontol A Biol Sci Med Sci ; 73(7): 853-863, 2018 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-29905772

RESUMEN

Obesity has deleterious effects on cognitive function in the elderly adults. In mice, aging exacerbates obesity-induced oxidative stress, microvascular dysfunction, blood-brain barrier (BBB) disruption, and neuroinflammation, which compromise cognitive health. However, the specific mechanisms through which aging and obesity interact to remain elusive. Previously, we have shown that Nrf2 signaling plays a critical role in microvascular resilience to obesity and that aging is associated with progressive Nrf2 dysfunction, promoting microvascular impairment. To test the hypothesis that Nrf2 deficiency exacerbates cerebromicrovascular dysfunction induced by obesity Nrf2+/+ and Nrf2-/-, mice were fed an adipogenic high-fat diet (HFD). Nrf2 deficiency significantly exacerbated HFD-induced oxidative stress and cellular senescence, impairment of neurovascular coupling responses, BBB disruption, and microglia activation, mimicking the aging phenotype. Obesity in Nrf2-/- mice elicited complex alterations in the amyloidogenic gene expression profile, including upregulation of amyloid precursor protein. Nrf2 deficiency and obesity additively reduced long-term potentiation in the CA1 area of the hippocampus. Collectively, Nrf2 dysfunction exacerbates the deleterious effects of obesity, compromising cerebromicrovascular and brain health by impairing neurovascular coupling mechanisms, BBB integrity and synaptic function and promoting neuroinflammation. These results support a possible role for age-related Nrf2 dysfunction in the pathogenesis of vascular cognitive impairment and Alzheimer's disease.


Asunto(s)
Envejecimiento/metabolismo , Factor 2 Relacionado con NF-E2/deficiencia , Obesidad/metabolismo , Envejecimiento/genética , Envejecimiento/psicología , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/metabolismo , Proteínas Amiloidogénicas/genética , Animales , Barrera Hematoencefálica/fisiopatología , Disfunción Cognitiva/metabolismo , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Expresión Génica , Hipocampo/metabolismo , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/fisiología , Plasticidad Neuronal , Obesidad/fisiopatología , Estrés Oxidativo , Fenotipo
4.
Geroscience ; 39(4): 385-406, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28664509

RESUMEN

Strong epidemiological and experimental evidence indicates that hypertension has detrimental effects on the cerebral microcirculation and thereby promotes accelerated brain aging. Hypertension is an independent risk factor for both vascular cognitive impairment (VCI) and Alzheimer's disease (AD). However, the pathophysiological link between hypertension-induced cerebromicrovascular injury (e.g., blood-brain barrier disruption, increased microvascular oxidative stress, and inflammation) and cognitive decline remains elusive. The present study was designed to characterize neuronal functional and morphological alterations induced by chronic hypertension and compare them to those induced by aging. To achieve that goal, we induced hypertension in young C57BL/6 mice by chronic (4 weeks) infusion of angiotensin II. We found that long-term potentiation (LTP) of performant path synapses following high-frequency stimulation of afferent fibers was decreased in hippocampal slices obtained from hypertensive mice, mimicking the aging phenotype. Hypertension and advanced age were associated with comparable decline in synaptic density in the stratum radiatum of the mouse hippocampus. Hypertension, similar to aging, was associated with changes in mRNA expression of several genes involved in regulation of neuronal function, including down-regulation of Bdnf, Homer1, and Dlg4, which may have a role in impaired synaptic plasticity. Collectively, hypertension impairs synaptic plasticity, reduces synaptic density, and promotes dysregulation of genes involved in synaptic function in the mouse hippocampus mimicking the aging phenotype. These hypertension-induced neuronal alterations may impair establishment of memories in the hippocampus and contribute to the pathogenesis and clinical manifestation of both vascular cognitive impairment (VCI) and Alzheimer's disease (AD).

5.
Age (Dordr) ; 38(4): 273-289, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27613724

RESUMEN

Strong epidemiological and experimental evidence indicate that both age and hypertension lead to significant functional and structural impairment of the cerebral microcirculation, predisposing to the development of vascular cognitive impairment (VCI) and Alzheimer's disease. Preclinical studies establish a causal link between cognitive decline and microvascular rarefaction in the hippocampus, an area of brain important for learning and memory. Age-related decline in circulating IGF-1 levels results in functional impairment of the cerebral microvessels; however, the mechanistic role of IGF-1 deficiency in impaired hippocampal microvascularization remains elusive. The present study was designed to characterize the additive/synergistic effects of IGF-1 deficiency and hypertension on microvascular density and expression of genes involved in angiogenesis and microvascular regression in the hippocampus. To achieve that goal, we induced hypertension in control and IGF-1 deficient mice (Igf1 f/f  + TBG-Cre-AAV8) by chronic infusion of angiotensin II. We found that circulating IGF-1 deficiency is associated with decreased microvascular density and exacerbates hypertension-induced microvascular rarefaction both in the hippocampus and the neocortex. The anti-angiogenic hippocampal gene expression signature observed in hypertensive IGF-1 deficient mice in the present study provides important clues for subsequent studies to elucidate mechanisms by which hypertension may contribute to the pathogenesis and clinical manifestation of VCI. In conclusion, adult-onset, isolated endocrine IGF-1 deficiency exerts deleterious effects on the cerebral microcirculation, leading to a significant decline in cortical and hippocampal capillarity and exacerbating hypertension-induced cerebromicrovascular rarefaction. The morphological impairment of the cerebral microvasculature induced by IGF-1 deficiency and hypertension reported here, in combination with neurovascular uncoupling, increased blood-brain barrier disruption and neuroinflammation reported in previous studies likely contribute to the pathogenesis of vascular cognitive impairment in elderly hypertensive humans.


Asunto(s)
Envejecimiento/metabolismo , Hipocampo/irrigación sanguínea , Hipertensión/complicaciones , Factor I del Crecimiento Similar a la Insulina/deficiencia , Rarefacción Microvascular/patología , Neocórtex/irrigación sanguínea , Envejecimiento/patología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Angiotensina II/efectos adversos , Angiotensina II/metabolismo , Animales , Biomarcadores/sangre , Barrera Hematoencefálica/metabolismo , Disfunción Cognitiva/fisiopatología , Expresión Génica , Humanos , Factor I del Crecimiento Similar a la Insulina/análisis , Factor I del Crecimiento Similar a la Insulina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Rarefacción Microvascular/etiología , ARN Mensajero/metabolismo
6.
Aging (Albany NY) ; 8(5): 899-916, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27070252

RESUMEN

Previous studies have shown positive effects of long-term resveratrol (RSV) supplementation in preventing pancreatic beta cell dysfunction, arterial stiffening and metabolic decline induced by high-fat/high-sugar (HFS) diet in nonhuman primates. Here, the analysis was extended to examine whether RSV may reduce dietary stress toxicity in the cerebral cortex of the same cohort of treated animals. Middle-aged male rhesus monkeys were fed for 2 years with HFS alone or combined with RSV, after which whole-genome microarray analysis of cerebral cortex tissue was carried out along with ELISA, immunofluorescence, and biochemical analyses to examine markers of vascular health and inflammation in the cerebral cortices. A number of genes and pathways that were differentially modulated in these dietary interventions indicated an exacerbation of neuroinflammation (e.g., oxidative stress markers, apoptosis, NF-κB activation) in HFS-fed animals and protection by RSV treatment. The decreased expression of mitochondrial aldehyde dehydrogenase 2, dysregulation in endothelial nitric oxide synthase, and reduced capillary density induced by HFS stress were rescued by RSV supplementation. Our results suggest that long-term RSV treatment confers neuroprotection against cerebral vascular dysfunction during nutrient stress.


Asunto(s)
Corteza Cerebral/efectos de los fármacos , Dieta Alta en Grasa , Sacarosa en la Dieta , Fármacos Neuroprotectores/farmacología , Estilbenos/farmacología , Animales , Apoptosis/efectos de los fármacos , Corteza Cerebral/metabolismo , Perfilación de la Expresión Génica , Inflamación/metabolismo , Macaca mulatta , Masculino , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Estrés Oxidativo/efectos de los fármacos , Resveratrol
7.
Am J Physiol Heart Circ Physiol ; 309(11): H1837-45, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26453330

RESUMEN

Impairment of moment-to-moment adjustment of cerebral blood flow (CBF) via neurovascular coupling is thought to play a critical role in the genesis of cognitive impairment associated with aging and pathological conditions associated with accelerated cerebromicrovascular aging (e.g., hypertension, obesity). Although previous studies demonstrate that endothelial dysfunction plays a critical role in neurovascular uncoupling in these conditions, the role of endothelial NO mediation in neurovascular coupling responses is not well understood. To establish the link between endothelial function and functional hyperemia, neurovascular coupling responses were studied in mutant mice overexpressing or deficient in endothelial NO synthase (eNOS), and the role of P2Y1 receptors in purinergic glioendothelial coupling was assessed. We found that genetic depletion of eNOS (eNOS(-/-)) and pharmacological inhibition of NO synthesis significantly decreased the CBF responses in the somatosensory cortex evoked by whisker stimulation and by administration of ATP. Overexpression of eNOS enhanced NO mediation of functional hyperemia. In control mice, the selective and potent P2Y1 receptor antagonist MRS2179 attenuated both whisker stimulation-induced and ATP-mediated CBF responses, whereas, in eNOS(-/-) mice, the inhibitory effects of MRS2179 were blunted. Collectively, our findings provide additional evidence for purinergic glio-endothelial coupling during neuronal activity, highlighting the role of ATP-mediated activation of eNOS via P2Y1 receptors in functional hyperemia.


Asunto(s)
Astrocitos/enzimología , Comunicación Celular , Células Endoteliales/enzimología , Hiperemia/enzimología , Microcirculación , Acoplamiento Neurovascular , Óxido Nítrico Sintasa de Tipo III/metabolismo , Receptores Purinérgicos P2Y1/metabolismo , Corteza Somatosensorial/enzimología , Animales , Comunicación Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Hemodinámica , Homeostasis , Hiperemia/genética , Hiperemia/fisiopatología , Mecanotransducción Celular , Ratones Endogámicos C57BL , Ratones Noqueados , Microcirculación/efectos de los fármacos , Acoplamiento Neurovascular/efectos de los fármacos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo III/deficiencia , Óxido Nítrico Sintasa de Tipo III/genética , Agonistas del Receptor Purinérgico P2Y/farmacología , Antagonistas del Receptor Purinérgico P2Y/farmacología , Receptores Purinérgicos P2Y1/efectos de los fármacos , Corteza Somatosensorial/irrigación sanguínea , Corteza Somatosensorial/efectos de los fármacos , Corteza Somatosensorial/fisiopatología , Vibrisas/inervación
8.
Aging Cell ; 14(6): 1034-44, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26172407

RESUMEN

Aging is associated with marked deficiency in circulating IGF-1, which has been shown to contribute to age-related cognitive decline. Impairment of moment-to-moment adjustment of cerebral blood flow (CBF) via neurovascular coupling is thought to play a critical role in the genesis of age-related cognitive impairment. To establish the link between IGF-1 deficiency and cerebromicrovascular impairment, neurovascular coupling mechanisms were studied in a novel mouse model of IGF-1 deficiency (Igf1(f/f) -TBG-Cre-AAV8) and accelerated vascular aging. We found that IGF-1-deficient mice exhibit neurovascular uncoupling and show a deficit in hippocampal-dependent spatial memory test, mimicking the aging phenotype. IGF-1 deficiency significantly impaired cerebromicrovascular endothelial function decreasing NO mediation of neurovascular coupling. IGF-1 deficiency also impaired glutamate-mediated CBF responses, likely due to dysregulation of astrocytic expression of metabotropic glutamate receptors and impairing mediation of CBF responses by eicosanoid gliotransmitters. Collectively, we demonstrate that IGF-1 deficiency promotes cerebromicrovascular dysfunction and neurovascular uncoupling mimicking the aging phenotype, which are likely to contribute to cognitive impairment.


Asunto(s)
Envejecimiento/genética , Circulación Cerebrovascular/fisiología , Trastornos del Conocimiento/genética , Factor I del Crecimiento Similar a la Insulina/genética , Acoplamiento Neurovascular/genética , Envejecimiento/fisiología , Animales , Encéfalo/fisiología , Trastornos del Conocimiento/fisiopatología , Modelos Animales de Enfermedad , Factor I del Crecimiento Similar a la Insulina/deficiencia , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microvasos/fisiología , Óxido Nítrico/biosíntesis , Memoria Espacial/fisiología
9.
J Cereb Blood Flow Metab ; 35(11): 1871-81, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26174328

RESUMEN

There is increasing evidence that vascular risk factors, including aging, hypertension, diabetes mellitus, and obesity, promote cognitive impairment; however, the underlying mechanisms remain obscure. Cerebral blood flow (CBF) is adjusted to neuronal activity via neurovascular coupling (NVC) and this mechanism is known to be impaired in the aforementioned pathophysiologic conditions. To establish a direct relationship between impaired NVC and cognitive decline, we induced neurovascular uncoupling pharmacologically in mice by inhibiting the synthesis of vasodilator mediators involved in NVC. Treatment of mice with the epoxygenase inhibitor N-(methylsulfonyl)-2-(2-propynyloxy)-benzenehexanamide (MSPPOH), the NO synthase inhibitor l-NG-Nitroarginine methyl ester (L-NAME), and the COX inhibitor indomethacin decreased NVC by over 60% mimicking the aging phenotype, which was associated with significantly impaired spatial working memory (Y-maze), recognition memory (Novel object recognition), and impairment in motor coordination (Rotarod). Blood pressure (tail cuff) and basal cerebral perfusion (arterial spin labeling perfusion MRI) were unaffected. Thus, selective experimental disruption of NVC is associated with significant impairment of cognitive and sensorimotor function, recapitulating neurologic symptoms and signs observed in brain aging and pathophysiologic conditions associated with accelerated cerebromicrovascular aging.


Asunto(s)
Trastornos del Conocimiento/inducido químicamente , Trastornos del Conocimiento/psicología , Acoplamiento Neurovascular/efectos de los fármacos , Animales , Presión Sanguínea/efectos de los fármacos , Circulación Cerebrovascular/efectos de los fármacos , Inhibidores de la Ciclooxigenasa/farmacología , Citocromo P-450 CYP2J2 , Sistema Enzimático del Citocromo P-450/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Potenciales Evocados Somatosensoriales/efectos de los fármacos , Marcha/efectos de los fármacos , Fuerza de la Mano , Indometacina/farmacología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa de Tipo I/antagonistas & inhibidores , Equilibrio Postural/efectos de los fármacos , Reconocimiento en Psicología/efectos de los fármacos
10.
Aging Cell ; 14(3): 400-8, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25677910

RESUMEN

Recent studies demonstrate that aging exacerbates hypertension-induced cognitive decline, but the specific age-related mechanisms remain elusive. Cerebral microhemorrhages (CMHs) are associated with rupture of small intracerebral vessels and are thought to progressively impair neuronal function. To determine whether aging exacerbates hypertension-induced CMHs young (3 months) and aged (24 months) mice were treated with angiotensin II plus L-NAME. We found that the same level of hypertension leads to significantly earlier onset and increased incidence of CMHs in aged mice than in young mice, as shown by neurological examination, gait analysis, and histological assessment of CMHs in serial brain sections. Hypertension-induced cerebrovascular oxidative stress and redox-sensitive activation of matrix metalloproteinases (MMPs) were increased in aging. Treatment of aged mice with resveratrol significantly attenuated hypertension-induced oxidative stress, inhibited vascular MMP activation, significantly delayed the onset, and reduced the incidence of CMHs. Collectively, aging promotes CMHs in mice likely by exacerbating hypertension-induced oxidative stress and MMP activation. Therapeutic strategies that reduce microvascular oxidative stress and MMP activation may be useful for the prevention of CMHs, protecting neurocognitive function in high-risk elderly patients.


Asunto(s)
Envejecimiento , Hipertensión/complicaciones , Hipertensión/tratamiento farmacológico , Hemorragias Intracraneales/etiología , Estilbenos/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Hemorragias Intracraneales/tratamiento farmacológico , Hemorragias Intracraneales/prevención & control , Masculino , Ratones Endogámicos C57BL , Oxidación-Reducción/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Resveratrol
11.
J Cereb Blood Flow Metab ; 35(4): 527-30, 2015 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-25605292

RESUMEN

Stability of myogenic tone in middle cerebral arteries (MCA) is essential for adequate control over penetration of pressure waves into the distal portion of the cerebral microcirculation. Because the increased pulse pressure observed in advanced aging is associated with cerebromicrovascular injury, the effect of aging on myogenic response of mouse MCAs was determined. Aging did not affect the myogenic constriction in response to static increases in pressure, whereas it significantly impaired pulsatile pressure-induced myogenic tone. Impaired myogenic adaptation of MCAs to pulsatile pressure may allow high pressure to penetrate the distal portion of the cerebral microcirculation, contributing to microvascular damage.


Asunto(s)
Envejecimiento , Encéfalo/irrigación sanguínea , Arteria Cerebral Media/fisiología , Flujo Pulsátil , Vasoconstricción , Adaptación Fisiológica , Animales , Presión Sanguínea , Masculino , Ratones , Ratones Endogámicos C57BL , Arteria Cerebral Media/fisiopatología
12.
J Gerontol A Biol Sci Med Sci ; 70(11): 1355-9, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25631392

RESUMEN

Epidemiological studies demonstrate that in addition to the increased prevalence of hypertension in old patients, the deleterious cerebrovascular effects of hypertension (including atherosclerosis, stroke, and vascular cognitive impairment) are also exacerbated in elderly individuals. The cellular mechanisms by which aging and hypertension interact to promote cerebrovascular pathologies are not well understood. To test the hypothesis that aging exacerbates high pressure-induced mitochondrial oxidative stress, we exposed isolated segments of the middle cerebral arteries of young (3 months) and aged (24 months) C57BL/6 mice to 60 or 140 mmHg intraluminal pressure and assessed changes in mitochondrial reactive oxygen species production using a mitochondria-targeted redox-sensitive fluorescent indicator dye (MitoSox) by confocal microscopy. Perinuclear MitoSox fluorescence was significantly stronger in high pressure-exposed middle cerebral arteries compared with middle cerebral arteries of the same animals exposed to 60 mmHg, indicating that high pressure increases mitochondrial reactive oxygen species production in the smooth muscle cells of cerebral arteries. Comparison of young and aged middle cerebral arteries showed that aging exacerbates high pressure-induced mitochondrial reactive oxygen species production in cerebral arteries. We propose that increased mechanosensitive mitochondrial oxidative stress may potentially exacerbate cerebrovascular injury and vascular inflammation in aging.


Asunto(s)
Arterias Cerebrales/metabolismo , Hipertensión/metabolismo , Mitocondrias/fisiología , Estrés Oxidativo/fisiología , Factores de Edad , Animales , Arterias Cerebrales/fisiopatología , Modelos Animales de Enfermedad , Hipertensión/etiología , Hipertensión/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos del Músculo Liso/fisiología , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Técnicas de Cultivo de Tejidos
13.
J Gerontol A Biol Sci Med Sci ; 70(6): 665-74, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25136000

RESUMEN

Aging impairs angiogenic capacity of cerebromicrovascular endothelial cells (CMVECs) promoting microvascular rarefaction, but the underlying mechanisms remain elusive. PACAP is an evolutionarily conserved neuropeptide secreted by endothelial cells and neurons, which confers important antiaging effects. To test the hypothesis that age-related changes in autocrine PACAP signaling contributes to dysregulation of endothelial angiogenic capacity, primary CMVECs were isolated from 3-month-old (young) and 24-month-old (aged) Fischer 344 x Brown Norway rats. In aged CMVECs, expression of PACAP was decreased, which was associated with impaired capacity to form capillary-like structures, impaired adhesiveness to collagen (assessed using electric cell-substrate impedance sensing [ECIS] technology), and increased apoptosis (caspase3 activity) when compared with young cells. Overexpression of PACAP in aged CMVECs resulted in increased formation of capillary-like structures, whereas it did not affect cell adhesion. Treatment with recombinant PACAP also significantly increased endothelial tube formation and inhibited apoptosis in aged CMVECs. In young CMVECs shRNA knockdown of autocrine PACAP expression significantly impaired tube formation capacity, mimicking the aging phenotype. Cellular and mitochondrial reactive oxygen species production (dihydroethidium and MitoSox fluorescence, respectively) were increased in aged CMVECs and were unaffected by PACAP. Collectively, PACAP exerts proangiogenic effects and age-related dysregulation of autocrine PACAP signaling may contribute to impaired angiogenic capacity of CMVECs in aging.


Asunto(s)
Envejecimiento/fisiología , Encéfalo/irrigación sanguínea , Células Endoteliales/metabolismo , Neovascularización Fisiológica , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Animales , Apoptosis , Caspasa 3/metabolismo , Adhesión Celular , Movimiento Celular , Células Cultivadas , Regulación hacia Abajo , Células Endoteliales/patología , Técnicas de Silenciamiento del Gen , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/genética , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas F344 , Especies Reactivas de Oxígeno/metabolismo , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/genética , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
14.
J Gerontol A Biol Sci Med Sci ; 70(3): 303-13, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24642904

RESUMEN

Resveratrol (3,4',5-trihydroxystilbene) is a plant-derived polyphenolic trans-stilbenoid, which exerts multifaceted antiaging effects. Here, we propose a novel delivery system for resveratrol, which significantly increases its cellular uptake into aged cells. Combination of resveratrol with a positively charged lipid component to "conventional" liposomes converts these lipid vesicles to a robust fusogenic system. To study their cellular uptake and cellular effects, we treated primary cerebromicrovascular endothelial cells isolated from aged F344xBN rats with resveratrol encapsulated in fusogenic liposomes (FL-RSV). To demonstrate effective cellular uptake of FL-RSV, accumulation of the lipophilic tracer dye, DiR, and resveratrol in cerebromicrovascular endothelial cells was confirmed using flow cytometry and confocal microscopy and high-performance liquid chromatography electrochemical detection. Treatment of aged cerebromicrovascular endothelial cells with FL-RSV activated Nrf2 (assessed with a reporter gene assay), significantly decreased cellular production of reactive oxygen species (assessed by a flow cytometry-based H2DCFDA fluorescence method), and inhibited apoptosis. Taken together, encapsulation of resveratrol into novel fusogenic liposomes significantly enhances the delivery of resveratrol into aged cells, which subsequently results in rapid activation of cellular Nrf2-driven antioxidant defense mechanisms. Our studies provide proof-of-concept for the development of a novel, translationally relevant interventional strategy for prevention and/or control of oxidative stress-related pathophysiological conditions in aging.


Asunto(s)
Antioxidantes/farmacología , Encéfalo/patología , Células Endoteliales/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo/efectos de los fármacos , Estilbenos/farmacología , Animales , Encéfalo/irrigación sanguínea , Encéfalo/metabolismo , Técnicas de Cultivo de Célula , Senescencia Celular/efectos de los fármacos , Células Endoteliales/metabolismo , Liposomas , Masculino , Vehículos Farmacéuticos , Ratas , Ratas Endogámicas BN , Ratas Endogámicas F344 , Especies Reactivas de Oxígeno/metabolismo , Resveratrol
15.
J Cereb Blood Flow Metab ; 34(12): 1887-97, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25248835

RESUMEN

Aging impairs autoregulatory protection in the brain, exacerbating hypertension-induced cerebromicrovascular injury, neuroinflammation, and development of vascular cognitive impairment. Despite the importance of the age-related decline in circulating insulin-like growth factor-1 (IGF-1) levels in cerebrovascular aging, the effects of IGF-1 deficiency on functional adaptation of cerebral arteries to high blood pressure remain elusive. To determine whether IGF-1 deficiency impairs autoregulatory protection, hypertension was induced in control and IGF-1-deficient mice (Igf1(f/f)+TBG-iCre-AAV8) by chronic infusion of angiotensin-II. In hypertensive control mice, cerebral blood flow (CBF) autoregulation was extended to higher pressure values and the pressure-induced tone of middle cerebral arteries (MCAs) was increased. In hypertensive IGF-1-deficient mice, autoregulation was markedly disrupted, and MCAs did not show adaptive increases in myogenic tone. In control mice, the mechanism of adaptation to hypertension involved upregulation of TRPC channels in MCAs and this mechanism was impaired in hypertensive IGF-1-deficient mice. Likely downstream consequences of cerebrovascular autoregulatory dysfunction in hypertensive IGF-1-deficient mice included exacerbated disruption of the blood-brain barrier and neuroinflammation (microglia activation and upregulation of proinflammatory cytokines and chemokines), which were associated with impaired hippocampal cognitive function. Collectively, IGF-1 deficiency impairs autoregulatory protection in the brain of hypertensive mice, potentially exacerbating cerebromicrovascular injury and neuroinflammation mimicking the aging phenotype.


Asunto(s)
Circulación Cerebrovascular/fisiología , Homeostasis/fisiología , Hipertensión/metabolismo , Hipertensión/fisiopatología , Factor I del Crecimiento Similar a la Insulina/genética , Envejecimiento/fisiología , Angiotensina II/farmacología , Animales , Biomarcadores/sangre , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/fisiología , Circulación Cerebrovascular/efectos de los fármacos , Demencia/inmunología , Demencia/metabolismo , Demencia/fisiopatología , Modelos Animales de Enfermedad , Encefalitis/inmunología , Encefalitis/metabolismo , Encefalitis/fisiopatología , Hipocampo/inmunología , Hipocampo/fisiopatología , Hipertensión/inmunología , Factor I del Crecimiento Similar a la Insulina/deficiencia , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/inmunología , Arteria Cerebral Media/fisiopatología , Canales Catiónicos TRPC/metabolismo , Vasoconstrictores/farmacología
16.
Am J Physiol Heart Circ Physiol ; 307(3): H292-306, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24906921

RESUMEN

In rodents, moderate caloric restriction (CR) without malnutrition exerts significant cerebrovascular protective effects, improving cortical microvascular density and endothelium-dependent vasodilation, but the underlying cellular mechanisms remain elusive. To elucidate the persisting effects of CR on cerebromicrovascular endothelial cells (CMVECs), primary CMVECs were isolated from young (3 mo old) and aged (24 mo old) ad libitum-fed and aged CR F344xBN rats. We found an age-related increase in cellular and mitochondrial oxidative stress, which is prevented by CR. Expression and transcriptional activity of Nrf2 are both significantly reduced in aged CMVECs, whereas CR prevents age-related Nrf2 dysfunction. Expression of miR-144 was upregulated in aged CMVECs, and overexpression of miR-144 significantly decreased expression of Nrf2 in cells derived from both young animals and aged CR rats. Overexpression of a miR-144 antagomir in aged CMVECs significantly decreases expression of miR-144 and upregulates Nrf2. We found that CR prevents age-related impairment of angiogenic processes, including cell proliferation, adhesion to collagen, and formation of capillary-like structures and inhibits apoptosis in CMVECs. CR also exerts significant anti-inflammatory effects, preventing age-related increases in the transcriptional activity of NF-κB and age-associated pro-inflammatory shift in the endothelial secretome. Characterization of CR-induced changes in miRNA expression suggests that they likely affect several critical functions in endothelial cell homeostasis. The predicted regulatory effects of CR-related differentially expressed miRNAs in aged CMVECs are consistent with the anti-aging endothelial effects of CR observed in vivo. Collectively, we find that CR confers persisting anti-oxidative, pro-angiogenic, and anti-inflammatory cellular effects, preserving a youthful phenotype in rat cerebromicrovascular endothelial cells, suggesting that through these effects CR may improve cerebrovascular function and prevent vascular cognitive impairment.


Asunto(s)
Envejecimiento/metabolismo , Encéfalo/irrigación sanguínea , Restricción Calórica , Células Endoteliales/metabolismo , Perfilación de la Expresión Génica , Inflamación/prevención & control , MicroARNs/metabolismo , Microvasos/metabolismo , Neovascularización Fisiológica , Estrés Oxidativo , Factores de Edad , Envejecimiento/genética , Envejecimiento/inmunología , Animales , Células Cultivadas , Cruzamientos Genéticos , Células Endoteliales/inmunología , Regulación de la Expresión Génica , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Mediadores de Inflamación/metabolismo , Masculino , MicroARNs/genética , Microvasos/inmunología , Mitocondrias/metabolismo , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Fenotipo , Ratas , Ratas Endogámicas BN , Ratas Endogámicas F344 , Transcripción Genética , Transfección
17.
J Gerontol A Biol Sci Med Sci ; 69(11): 1339-52, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24895269

RESUMEN

Epidemiological studies show that obesity has deleterious effects on the brain and cognitive function in the elderly population. However, the specific mechanisms through which aging and obesity interact to promote cognitive decline remain unclear. To test the hypothesis that aging exacerbates obesity-induced cerebromicrovascular impairment, we compared young (7 months) and aged (24 months) high-fat diet-fed obese C57BL/6 mice. We found that aging exacerbates the obesity-induced decline in microvascular density both in the hippocampus and in the cortex. The extent of hippocampal microvascular rarefaction and the extent of impairment of hippocampal-dependent cognitive function positively correlate. Aging exacerbates obesity-induced loss of pericyte coverage on cerebral microvessels and alters hippocampal angiogenic gene expression signature, which likely contributes to microvascular rarefaction. Aging also exacerbates obesity-induced oxidative stress and induction of NADPH oxidase and impairs cerebral blood flow responses to whisker stimulation. Collectively, obesity exerts deleterious cerebrovascular effects in aged mice, promoting cerebromicrovascular rarefaction and neurovascular uncoupling. The morphological and functional impairment of the cerebral microvasculature in association with increased blood-brain barrier disruption and neuroinflammation (Tucsek Z, Toth P, Sosnowsk D, et al. Obesity in aging exacerbates blood-brain barrier disruption, neuroinflammation and oxidative stress in the mouse hippocampus: effects on expression of genes involved in beta-amyloid generation and Alzheimer's disease. J Gerontol Biol Med Sci. 2013. In press, PMID: 24269929) likely contribute to obesity-induced cognitive decline in aging.


Asunto(s)
Envejecimiento/patología , Envejecimiento/psicología , Encéfalo/irrigación sanguínea , Encéfalo/patología , Trastornos del Conocimiento/etiología , Obesidad/complicaciones , Envejecimiento/genética , Animales , Barrera Hematoencefálica/patología , Barrera Hematoencefálica/fisiopatología , Encéfalo/fisiopatología , Circulación Cerebrovascular , Trastornos del Conocimiento/patología , Trastornos del Conocimiento/psicología , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Fuerza de la Mano/fisiología , Hipocampo/irrigación sanguínea , Hipocampo/patología , Hipocampo/fisiopatología , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Microvasos/patología , Neovascularización Fisiológica/genética , Obesidad/patología , Obesidad/psicología , Estrés Oxidativo , Pericitos/patología , Transcriptoma
18.
J Gerontol A Biol Sci Med Sci ; 69(10): 1212-26, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24269929

RESUMEN

There is growing evidence that obesity has deleterious effects on the brain and cognitive function in the elderly population. However, the specific mechanisms through which aging and obesity interact to promote cognitive decline remain unclear. To test the hypothesis that aging exacerbates obesity-induced cerebromicrovascular damage and neuroinflammation, we compared young (7 months) and aged (24 months) high fat diet-fed obese C57BL/6 mice. Aging exacerbated obesity-induced systemic inflammation and blood-brain barrier disruption, as indicated by the increased circulating levels of proinflammatory cytokines and increased presence of extravasated immunoglobulin G in the hippocampus, respectively. Obesity-induced blood-brain barrier damage was associated with microglia activation, upregulation of activating Fc-gamma receptors and proinflammatory cytokines, and increased oxidative stress. Treatment of cultured primary microglia with sera derived from aged obese mice resulted in significantly more pronounced microglia activation and oxidative stress, as compared with treatment with young sera. Serum-induced activation and oxidative stress were also exacerbated in primary microglia derived from aged animals. Hippocampal expression of genes involved in regulation of the cellular amyloid precursor protein-dependent signaling pathways, beta-amyloid generation, and the pathogenesis of tauopathy were largely unaffected by obesity in aged mice. Collectively, obesity in aging is associated with a heightened state of systemic inflammation, which exacerbates blood-brain barrier disruption. The resulting neuroinflammation and oxidative stress in the mouse hippocampus likely contribute to the significant cognitive decline observed in aged obese animals.


Asunto(s)
Envejecimiento/metabolismo , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/biosíntesis , Barrera Hematoencefálica , Hipocampo/metabolismo , Obesidad/complicaciones , Estrés Oxidativo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Arterias Cerebrales/fisiología , Dieta Alta en Grasa , Homeostasis , Inflamación/etiología , Masculino , Ratones , Ratones Endogámicos C57BL
19.
Am J Physiol Heart Circ Physiol ; 306(3): H299-308, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24322615

RESUMEN

Moment-to-moment adjustment of cerebral blood flow (CBF) to neuronal activity via neurovascular coupling is essential for the maintenance of normal neuronal function. Increased oxidative stress that occurs with aging was shown to impair neurovascular coupling, which likely contributes to a significant age-related decline in higher cortical function, increasing the risk for vascular cognitive impairment. Resveratrol is a polyphenolic compound that exerts significant antiaging protective effects in large vessels, but its effects on the cerebromicrovasculature remain poorly defined. The present study was undertaken to investigate the capacity of resveratrol to improve neurovascular coupling in aging. In aged (24-mo-old) C57BL/6 mice N(ω)-nitro-l-arginine methyl ester-sensitive, nitric oxide-mediated CBF responses to whisker stimulation and to the endothelium-dependent dilator acethylcholine (ACh) were impaired compared with those in young (3-mo-old) mice. Treatment of aged mice with resveratrol rescued neurovascular coupling and ACh-induced responses, which was associated with downregulation of cortical expression of NADPH oxidase and decreased levels of biomarkers of oxidative/nitrative stress (3-nitrotyrosine, 8-isoprostanes). Resveratrol also attenuated age-related increases in reactive oxygen species (ROS) production in cultured cerebromicrovascular endothelial cells (DCF fluorescence, flow cytometry). In conclusion, treatment with resveratrol rescues cortical neurovascular coupling responses to increased neuronal activity in aged mice, likely by restoring cerebromicrovascular endothelial function via downregulation of NADPH oxidase-derived ROS production. Beneficial cerebromicrovascular effects of resveratrol may contribute to its protective effects on cognitive function in aging.


Asunto(s)
Envejecimiento/fisiología , Cerebro/irrigación sanguínea , Endotelio Vascular/efectos de los fármacos , Microcirculación/efectos de los fármacos , Estilbenos/farmacología , Vasodilatación/efectos de los fármacos , Animales , Demencia Vascular/prevención & control , Endotelio Vascular/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Microcirculación/fisiología , NADPH Oxidasas/metabolismo , Estrés Oxidativo/fisiología , Especies Reactivas de Oxígeno/metabolismo , Resveratrol , Vasodilatadores/farmacología
20.
Am J Physiol Heart Circ Physiol ; 305(12): H1698-708, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24097425

RESUMEN

Hypertension in the elderly substantially increases the risk of stroke and vascular cognitive impairment in part due to an impaired functional adaptation of aged cerebral arteries to high blood pressure. To elucidate the mechanisms underlying impaired autoregulatory protection in aging, hypertension was induced in young (3 mo) and aged (24 mo) C57BL/6 mice by chronic infusion of angiotensin II and pressure-induced changes in smooth muscle cell (SMC) intracellular Ca(2+) concentration ([Ca(2+)]i) and myogenic constriction of middle cerebral arteries (MCA) were assessed. In MCAs from young hypertensive mice, pressure-induced increases in vascular SMC [Ca(2+)]i and myogenic tone were increased, and these adaptive responses were inhibited by the cytochrome P-450 ω-hydroxylase inhibitor HET0016 and the transient receptor potential (TRP) channel blocker SKF96365. Administration of 20- hydroxyeicosatetraenoic acid (HETE) increased SMC [Ca(2+)]i and constricted MCAs, and these responses were inhibited by SKF96365. MCAs from aged hypertensive mice did not show adaptive increases in pressure-induced calcium signal and myogenic tone and responses to HET0016 and SKF96365 were blunted. Inhibition of large-conductance Ca(2+)-activated K(+) (BK) channels by iberiotoxin enhanced SMC [Ca(2+)]i and myogenic constriction in MCAs of young normotensive animals, whereas it was without effect in MCAs of young hypertensive mice. Iberiotoxin did not restore myogenic adaptation in MCAs of aged hypertensive mice. Thus functional maladaptation of aged cerebral arteries to hypertension is due to the dysregulation of pressure-induced 20-HETE and TRP channel-mediated SMC calcium signaling, whereas overactivation of BK channels is unlikely to play a role in this phenomenon.


Asunto(s)
Señalización del Calcio/fisiología , Ácidos Hidroxieicosatetraenoicos/farmacología , Hipertensión/metabolismo , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/metabolismo , Arteria Cerebral Media/metabolismo , Canales Catiónicos TRPC/metabolismo , Vasoconstricción/fisiología , Envejecimiento/fisiología , Angiotensina II , Animales , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Hipertensión/inducido químicamente , Hipertensión/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Arteria Cerebral Media/efectos de los fármacos , Arteria Cerebral Media/fisiopatología , Contracción Muscular/efectos de los fármacos , Contracción Muscular/fisiología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/fisiopatología , Vasoconstricción/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA