Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Elife ; 92020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-33016877

RESUMEN

Cortical neural activity is coupled to local arterial diameter and blood flow. However, which neurons control the dynamics of cerebral arteries is not well understood. We dissected the cellular mechanisms controlling the basal diameter and evoked dilation in cortical arteries in awake, head-fixed mice. Locomotion drove robust arterial dilation, increases in gamma band power in the local field potential (LFP), and increases calcium signals in pyramidal and neuronal nitric oxide synthase (nNOS)-expressing neurons. Chemogenetic or pharmocological modulation of overall neural activity up or down caused corresponding increases or decreases in basal arterial diameter. Modulation of pyramidal neuron activity alone had little effect on basal or evoked arterial dilation, despite pronounced changes in the LFP. Modulation of the activity of nNOS-expressing neurons drove changes in the basal and evoked arterial diameter without corresponding changes in population neural activity.


Asunto(s)
Arterias Cerebrales/fisiopatología , Interneuronas/metabolismo , Óxido Nítrico Sintasa de Tipo I/genética , Corteza Somatosensorial/metabolismo , Animales , Arterias Cerebrales/metabolismo , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo I/metabolismo
3.
Fluids Barriers CNS ; 17(1): 52, 2020 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-32819402

RESUMEN

The brain lacks a conventional lymphatic system to remove metabolic waste. It has been proposed that directional fluid movement through the arteriolar paravascular space (PVS) promotes metabolite clearance. We performed simulations to examine if arteriolar pulsations and dilations can drive directional CSF flow in the PVS and found that arteriolar wall movements do not drive directional CSF flow. We propose an alternative method of metabolite clearance from the PVS, namely fluid exchange between the PVS and the subarachnoid space (SAS). In simulations with compliant brain tissue, arteriolar pulsations did not drive appreciable fluid exchange between the PVS and the SAS. However, when the arteriole dilated, as seen during functional hyperemia, there was a marked exchange of fluid. Simulations suggest that functional hyperemia may serve to increase metabolite clearance from the PVS. We measured blood vessels and brain tissue displacement simultaneously in awake, head-fixed mice using two-photon microscopy. These measurements showed that brain deforms in response to pressure changes in PVS, consistent with our simulations. Our results show that the deformability of the brain tissue needs to be accounted for when studying fluid flow and metabolite transport.


Asunto(s)
Encéfalo/irrigación sanguínea , Encéfalo/metabolismo , Líquido Cefalorraquídeo/metabolismo , Hiperemia/líquido cefalorraquídeo , Hiperemia/metabolismo , Animales , Arteriolas/metabolismo , Humanos , Modelos Neurológicos , Espacio Subaracnoideo/metabolismo
4.
Nat Neurosci ; 20(12): 1761-1769, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29184204

RESUMEN

Spontaneous fluctuations in hemodynamic signals in the absence of a task or overt stimulation are used to infer neural activity. We tested this coupling by simultaneously measuring neural activity and changes in cerebral blood volume (CBV) in the somatosensory cortex of awake, head-fixed mice during periods of true rest and during whisker stimulation and volitional whisking. We found that neurovascular coupling was similar across states and that large, spontaneous CBV changes in the absence of sensory input were driven by volitional whisker and body movements. Hemodynamic signals during periods of rest were weakly correlated with neural activity. Spontaneous fluctuations in CBV and vessel diameter persisted when local neural spiking and glutamatergic input were blocked, as well as during blockade of noradrenergic receptors, suggesting a non-neuronal origin for spontaneous CBV fluctuations. Spontaneous hemodynamic signals reflect a combination of behavior, local neural activity, and putatively non-neural processes.


Asunto(s)
Hemodinámica/fisiología , Descanso/fisiología , Animales , Conducta Animal/fisiología , Volumen Sanguíneo , Circulación Cerebrovascular/fisiología , Nervio Facial/fisiología , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Movimiento/fisiología , Fenómenos Fisiológicos del Sistema Nervioso , Estimulación Luminosa , Corteza Somatosensorial/irrigación sanguínea , Vibrisas/inervación , Vibrisas/fisiología
5.
J Transl Med ; 15(1): 55, 2017 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-28264687

RESUMEN

OBJECTIVE: Low to moderate inorganic arsenic (iAs) exposure is independently associated with cardiovascular disease (CVD), particularly for patients with diabetes mellitus (DM). The mechanism of increased CVD risk from iAs exposure in DM has not been adequately characterized. We evaluated whether increasing concentrations of glucose enhance the effects of iAs on platelet and megakaryocyte activity, key steps in atherothrombosis. METHODS: Healthy donor whole blood was prepared in a standard fashion and incubated with sodium arsenite in a range from 0 to 10 µM. iAs-induced platelet activation was assessed by platelet receptor CD62P (P-selectin) expression and monocyte-platelet and leukocyte-platelet aggregation (MPA and LPA, respectively) in the presence of increasing sodium arsenite and glucose concentrations. Megakaryocyte (Meg-01) cell adhesion and gene expression was assessed after incubation with or without iAs and increasing concentrations of D-glucose. RESULTS: Platelet activity markers increased significantly with 10 vs. 0 µM iAs (P < 0.05 for all) and with higher D-glucose concentrations. Platelet activity increased significantly following co incubation of 1 and 5 µM iAs concentrations with hyperglycemic D-glucose (P < 0.01 for both) but not after incubation with euglycemic D-glucose. Megakaryocyte adhesion was more pronounced after co incubation with iAs and hyperglycemic than euglycemic D-glucose, while gene expression increased significantly to iAs only after co incubation with hyperglycemic D-glucose. CONCLUSION: We demonstrate that glucose concentrations common in DM potentiate the effect of inorganic arsenic exposure on markers of platelet and megakaryocyte activity. Our results support recent observational cohort data that DM enhances the vasculotoxic effects of arsenic exposure, and suggest that activation of the platelet-megakaryocyte hemostatic axis is a pathway through which inorganic arsenic confers atherothrombotic risk, particularly for patients with DM.


Asunto(s)
Arsénico/farmacología , Plaquetas/metabolismo , Hiperglucemia/patología , Megacariocitos/metabolismo , Activación Plaquetaria/efectos de los fármacos , Plaquetas/efectos de los fármacos , Plaquetas/patología , Adhesión Celular/efectos de los fármacos , Citometría de Flujo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Megacariocitos/efectos de los fármacos , Megacariocitos/patología , FN-kappa B/genética , FN-kappa B/metabolismo , Selectina-P/metabolismo , Trombina/farmacología
6.
Blood ; 128(16): 2033-2042, 2016 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-27609643

RESUMEN

Platelet activity plays a major role in hemostasis with increased platelet activity likely contributing to the pathogenesis of atherothrombosis. We sought to identify associations between platelet activity variability and platelet-related genes in healthy controls. Transcriptional profiling of platelets revealed that WD-40 repeat domain 1 (WDR1), an enhancer of actin-depolymerizing factor activity, is downregulated in platelet messenger RNA (mRNA) from subjects with a hyperreactive platelet phenotype. We used the human megakaryoblastic cell line MEG-01 as an in vitro model for human megakaryocytes and platelets. Stimulation of MEG-01 with thrombin reduced levels of WDR1 transcripts and protein. WDR1 knockdown (KD) in MEG-01 cells increased adhesion and spreading in both the basal and activated states, increased F-actin content, and increased the basal intracellular calcium concentration. Platelet-like particles (PLPs) produced by WDR1 KD cells were fewer in number but larger than PLPs produced from unmodified MEG-01 cells, and had significantly increased adhesion in the basal state and upon thrombin activation. In contrast, WDR1 overexpression reversed the WDR1 KD phenotype of megakaryocytes and PLPs. To translate the clinical significance of these findings, WDR1 expression was measured in platelet RNA from subjects with established cardiovascular disease (n = 27) and age- and sex-matched controls (n = 10). The WDR1 mRNA and protein level was significantly lower in subjects with cardiovascular disease. These data suggest that WDR1 plays an important role in suppressing platelet activity, where it alters the actin cytoskeleton dynamics, and downregulation of WDR1 may contribute to the platelet-mediated pathogenesis of cardiovascular disease.


Asunto(s)
Aterosclerosis/metabolismo , Plaquetas/metabolismo , Citoesqueleto/metabolismo , Regulación de la Expresión Génica , Proteínas de Microfilamentos/biosíntesis , Adhesividad Plaquetaria , Adulto , Aterosclerosis/genética , Aterosclerosis/patología , Plaquetas/patología , Línea Celular , Citoesqueleto/genética , Citoesqueleto/patología , Femenino , Humanos , Masculino , Megacariocitos/metabolismo , Megacariocitos/patología , Proteínas de Microfilamentos/genética
8.
Inflammation ; 39(1): 182-189, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26318864

RESUMEN

The cardioprotective mechanisms of colchicine in patients with stable ischemic heart disease remain uncertain. We tested varying concentrations of colchicine on platelet activity in vitro and a clinically relevant 1.8-mg oral loading dose administered over 1 h in 10 healthy subjects. Data are shown as median [interquartile range]. Colchicine addition in vitro decreased light transmission platelet aggregation only at supratherapeutic concentrations but decreased monocyte- (MPA) and neutrophil-platelet aggregation (NPA) at therapeutic concentrations. Administration of 1.8 mg colchicine to healthy subjects had no significant effect on light transmission platelet aggregation but decreased the extent of MPA (28 % [22-57] to 22 % [19-31], p = 0.05) and NPA (19 % [16-59] to 15 % [11-30], p = 0.01), platelet surface expression of PAC-1 (370 mean fluorescence intensity (MFI) [328-555] to 333 MFI [232-407], p = 0.02) and P-selectin (351 MFI [269-492] to 279 [226-364], p = 0.03), and platelet adhesion to collagen (10.2 % [2.5-32.6] to 2.0 % [0.2-9.5], p = 0.09) 2 h post-administration. Thus, in clinically relevant concentrations, colchicine decreases expression of surface markers of platelet activity and inhibits leukocyte-platelet aggregation but does not inhibit homotypic platelet aggregation.


Asunto(s)
Plaquetas/metabolismo , Cardiotónicos/farmacología , Colchicina/farmacología , Monocitos/metabolismo , Neutrófilos/metabolismo , Agregación Plaquetaria/efectos de los fármacos , Adulto , Plaquetas/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Femenino , Voluntarios Sanos , Humanos , Inflamación/tratamiento farmacológico , Masculino , Monocitos/efectos de los fármacos , Isquemia Miocárdica/tratamiento farmacológico , Neutrófilos/efectos de los fármacos , Proyectos Piloto , Activación Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/fisiología , Estudios Prospectivos , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...