Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
Cereb Circ Cogn Behav ; 5: 100189, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37941765

RESUMEN

Although dementia research has been dominated by Alzheimer's disease (AD), most dementia in older people is now recognised to be due to mixed pathologies, usually combining vascular and AD brain pathology. Vascular cognitive impairment (VCI), which encompasses vascular dementia (VaD) is the second most common type of dementia. Models of VCI have been delayed by limited understanding of the underlying aetiology and pathogenesis. This review by a multidisciplinary, diverse (in terms of sex, geography and career stage), cross-institute team provides a perspective on limitations to current VCI models and recommendations for improving translation and reproducibility. We discuss reproducibility, clinical features of VCI and corresponding assessments in models, human pathology, bioinformatics approaches, and data sharing. We offer recommendations for future research, particularly focusing on small vessel disease as a main underpinning disorder.

2.
Commun Biol ; 6(1): 185, 2023 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-36797344

RESUMEN

Neurovascular coupling (NVC) is a mechanism that, amongst other known and latent critical functions, ensures activated brain regions are adequately supplied with oxygen and glucose. This biological phenomenon underpins non-invasive perfusion-related neuroimaging techniques and recent reports have implicated NVC impairment in several neurodegenerative disorders. Yet, much remains unknown regarding NVC in health and disease, and only recently has there been burgeoning recognition of a close interplay with brain thermodynamics. Accordingly, we developed a novel multi-modal approach to systematically modulate cortical temperature and interrogate the spatiotemporal dynamics of sensory-evoked NVC. We show that changes in cortical temperature profoundly and intricately modulate NVC, with low temperatures associated with diminished oxygen delivery, and high temperatures inducing a distinct vascular oscillation. These observations provide novel insights into the relationship between NVC and brain thermodynamics, with important implications for brain-temperature related therapies, functional biomarkers of elevated brain temperature, and in-vivo methods to study neurovascular coupling.


Asunto(s)
Encéfalo , Acoplamiento Neurovascular , Temperatura , Acoplamiento Neurovascular/fisiología , Reconocimiento en Psicología , Oxígeno
3.
J Biomech Eng ; 145(3)2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36445228

RESUMEN

Neurovascular coupling (NVC) is the ability to locally adjust vascular resistance as a function of neuronal activity. Recent experiments have illustrated that NVC is partially independent of metabolic signals. In addition, nitric oxide (NO) has been shown in some instances to provide an important mechanism in altering vascular resistance. An extension to the original model of NVC [1] has been developed to include the activation of both somatosensory neurons and GABAergic interneurons and to investigate the role of NO and the delicate balance of GABA and neuronal peptide enzymes (NPY) pathways. The numerical model is compared to murine experimental data that provides time-dependent profiles of oxy, de-oxy, and total-hemoglobin. The results indicate a delicate balance that exists between GABA and NPY when nNOS interneurons are activated mediated by NO. Whereas somatosensory neurons (producing potassium into the extracellular space) do not seem to be effected by the inhibition of NO. Further work will need to be done to investigate the role of NO when stimulation periods are increased substantially from the short pulses of 2 s as used in the above experiments.


Asunto(s)
Neuronas GABAérgicas , Acoplamiento Neurovascular , Animales , Ratones , Acoplamiento Neurovascular/fisiología , Interneuronas/metabolismo , Ácido gamma-Aminobutírico/metabolismo
4.
Sci Rep ; 12(1): 6236, 2022 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-35422473

RESUMEN

Investigating neurovascular coupling in awake rodents is becoming ever more popular due, in part, to our increasing knowledge of the profound impacts that anaesthesia can have upon brain physiology. Although awake imaging brings with it many advantages, we still do not fully understand how voluntary locomotion during imaging affects sensory-evoked haemodynamic responses. In this study we investigated how evoked haemodynamic responses can be affected by the amount and timing of locomotion. Using an awake imaging set up, we used 2D-Optical Imaging Spectroscopy (2D-OIS) to measure changes in cerebral haemodynamics within the sensory cortex of the brain during either 2 s whisker stimulation or spontaneous (no whisker stimulation) experiments, whilst animals could walk on a spherical treadmill. We show that locomotion alters haemodynamic responses. The amount and timing of locomotion relative to whisker stimulation is important, and can significantly impact sensory-evoked haemodynamic responses. If locomotion occurred before or during whisker stimulation, the amplitude of the stimulus-evoked haemodynamic response was significantly altered. Therefore, monitoring of locomotion during awake imaging is necessary to ensure that conclusions based on comparisons of evoked haemodynamic responses (e.g., between control and disease groups) are not confounded by the effects of locomotion.


Asunto(s)
Corteza Somatosensorial , Vigilia , Animales , Hemodinámica/fisiología , Locomoción , Ratones , Estimulación Física/métodos , Corteza Somatosensorial/fisiología , Vibrisas/fisiología , Vigilia/fisiología
5.
Elife ; 112022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-35014950

RESUMEN

Neurovascular coupling is a critical brain mechanism whereby changes to blood flow accompany localised neural activity. The breakdown of neurovascular coupling is linked to the development and progression of several neurological conditions including dementia. In this study, we examined cortical haemodynamics in mouse preparations that modelled Alzheimer's disease (J20-AD) and atherosclerosis (PCSK9-ATH) between 9 and 12 m of age. We report novel findings with atherosclerosis where neurovascular decline is characterised by significantly reduced blood volume, altered levels of oxyhaemoglobin and deoxyhaemoglobin, in addition to global neuroinflammation. In the comorbid mixed model (J20-PCSK9-MIX), we report a 3 x increase in hippocampal amyloid-beta plaques. A key finding was that cortical spreading depression (CSD) due to electrode insertion into the brain was worse in the diseased animals and led to a prolonged period of hypoxia. These findings suggest that systemic atherosclerosis can be detrimental to neurovascular health and that having cardiovascular comorbidities can exacerbate pre-existing Alzheimer's-related amyloid-plaques.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Aterosclerosis/fisiopatología , Acoplamiento Neurovascular/fisiología , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Aterosclerosis/sangre , Encéfalo/metabolismo , Circulación Cerebrovascular/fisiología , Depresión de Propagación Cortical , Modelos Animales de Enfermedad , Hemodinámica , Masculino , Ratones , Ratones Endogámicos C57BL
6.
Neuroimage ; 237: 118195, 2021 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-34038769

RESUMEN

Cerebral blood volume (CBV) has been shown to be a robust and important physiological parameter for quantitative interpretation of functional (f)MRI, capable of delivering highly localized mapping of neural activity. Indeed, with recent advances in ultra-high-field (≥7T) MRI hardware and associated sequence libraries, it has become possible to capture non-invasive CBV weighted fMRI signals across cortical layers. One of the most widely used approaches to achieve this (in humans) is through vascular-space-occupancy (VASO) fMRI. Unfortunately, the exact contrast mechanisms of layer-dependent VASO fMRI have not been validated for human fMRI and thus interpretation of such data is confounded. Here we validate the signal source of layer-dependent SS-SI VASO fMRI using multi-modal imaging in a rat model in response to neuronal activation (somatosensory cortex) and respiratory challenge (hypercapnia). In particular VASO derived CBV measures are directly compared to concurrent measures of total haemoglobin changes from high resolution intrinsic optical imaging spectroscopy (OIS). Quantified cortical layer profiling is demonstrated to be in agreement between VASO and contrast enhanced fMRI (using monocrystalline iron oxide nanoparticles, MION). Responses show high spatial localisation to layers of cortical processing independent of confounding large draining veins which can hamper BOLD fMRI studies, (depending on slice positioning). Thus, a cross species comparison is enabled using VASO as a common measure. We find increased VASO based CBV reactivity (3.1 ± 1.2 fold increase) in humans compared to rats. Together, our findings confirm that the VASO contrast is indeed a reliable estimate of layer-specific CBV changes. This validation study increases the neuronal interpretability of human layer-dependent VASO fMRI as an appropriate method in neuroscience application studies, in which the presence of large draining intracortical and pial veins limits neuroscientific inference with BOLD fMRI.


Asunto(s)
Volumen Sanguíneo Cerebral/fisiología , Neuroimagen Funcional/normas , Imagen por Resonancia Magnética/normas , Corteza Somatosensorial/diagnóstico por imagen , Percepción del Tacto/fisiología , Adulto , Animales , Estimulación Eléctrica , Femenino , Humanos , Masculino , Imagen Óptica , Estimulación Física , Ratas , Reproducibilidad de los Resultados
7.
Mech Ageing Dev ; 192: 111361, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32998028

RESUMEN

The use of animal models is fundamental to furthering our understanding of human disease mechanisms, as well as identifying potential therapeutic targets. Diseases of ageing often involve multiple body systems; however, multi-systemic features are not fully recapitulated in the many of the animal models available. Therefore, combining pre-clinical models to better reflect the multimorbidities observed at the clinical level is critical. This review will highlight some of the key pre-clinical experimental models associated with cardiovascular (atherosclerosis, coronary heart disease), cerebrovascular (stroke, vascular dementia), metabolic (obesity, type-2 diabetes mellitus) and neurological (amyotrophic lateral sclerosis, frontotemporal dementia, Parkinson's, epilepsy) diseases, and whether these models encompass known multimorbidities. In addition to this, we discuss established pre-clinical models that combine two or more conditions, within the context of dementia.


Asunto(s)
Envejecimiento/fisiología , Enfermedades Cardiovasculares , Modelos Animales de Enfermedad , Enfermedades Metabólicas , Enfermedades Neurodegenerativas , Animales , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/fisiopatología , Evaluación Preclínica de Medicamentos/métodos , Humanos , Enfermedades Metabólicas/tratamiento farmacológico , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/fisiopatología , Multimorbilidad , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/fisiopatología
8.
Brain Behav Immun Health ; 5: 100074, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32685933

RESUMEN

Brain health relies on a tightly regulated system known as neurovascular coupling whereby the cellular constituents of the neuro-glial-vascular unit (NGVU) regulate cerebral haemodynamics in accordance with brain metabolic demand. Disruption of neurovascular coupling impairs brain health and is associated with the development of a number for neurological conditions, including Alzheimer's disease. The NGVU is also a key site of action for neuroinflammatory responses and contributes to the transition of systemic inflammation to neuroinflammatory processes. Thus, systemic inflammatory challenges may cause a shift in NGVU operation towards prioritising neuroinflammatory action and thus altering neurovascular coupling and resultant cerebrovascular changes. To investigate this, rats were injected with lipopolysaccharide (LPS) (2 â€‹mg/kg) to induce a systemic inflammatory response, or vehicle, and brain haemodynamic responses to sensory and non-sensory (hypercapnia) stimuli were assessed in vivo using optical imaging techniques. Following imaging, animals were perfused and their brains extracted to histologically characterise components of the NGVU to determine the association between underlying cellular changes and in vivo blood flow regulation. LPS-treated animals showed changes in haemodynamic function and cerebrovascular dynamics 6 â€‹hours after LPS administration. Histological assessment identified a significant increase in astrogliosis, microgliosis and endothelial activation in LPS-treated animals. Our data shows that an acutely induced systemic inflammatory response is able to rapidly alter in vivo haemodynamic function and is associated with significant changes in the cellular constituents of the NGVU. We suggest that these effects are initially mediated by endothelial cells, which are directly exposed to the circulating inflammatory stimulus and have been implicated in regulating functional hyperaemia.

9.
Sci Rep ; 10(1): 7518, 2020 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-32371859

RESUMEN

Early impairments to neurovascular coupling have been proposed to be a key pathogenic factor in the onset and progression of Alzheimer's disease (AD). Studies have shown impaired neurovascular function in several mouse models of AD, including the J20-hAPP mouse. In this study, we aimed to investigate early neurovascular changes using wild-type (WT) controls and J20-hAPP mice at 6 months of age, by measuring cerebral haemodynamics and neural activity to physiological sensory stimulations. A thinned cranial window was prepared to allow access to cortical vasculature and imaged using 2D-optical imaging spectroscopy (2D-OIS). After chronic imaging sessions where the skull was intact, a terminal acute imaging session was performed where an electrode was inserted into the brain to record simultaneous neural activity. We found that cerebral haemodynamic changes were significantly enhanced in J20-hAPP mice compared with controls in response to physiological stimulations, potentially due to the significantly higher neural activity (hyperexcitability) seen in the J20-hAPP mice. Thus, neurovascular coupling remained preserved under a chronic imaging preparation. Further, under hyperoxia, the baseline blood volume and saturation of all vascular compartments in the brains of J20-hAPP mice were substantially enhanced compared to WT controls, but this effect disappeared under normoxic conditions. This study highlights novel findings not previously seen in the J20-hAPP mouse model, and may point towards a potential therapeutic strategy.


Asunto(s)
Enfermedad de Alzheimer/sangre , Precursor de Proteína beta-Amiloide/genética , Volumen Sanguíneo Cerebral , Hiperoxia/patología , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Hemodinámica , Heterocigoto , Hipocampo/metabolismo , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/metabolismo , Oxígeno/metabolismo
10.
J Cereb Blood Flow Metab ; 40(11): 2289-2303, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-31760864

RESUMEN

Impaired neurovascular coupling has been suggested as an early pathogenic factor in Alzheimer's disease (AD), which could serve as an early biomarker of cerebral pathology. We have established an anaesthetic regime to allow repeated measurements of neurovascular function over three months in the J20 mouse model of AD (J20-AD) and wild-type (WT) controls. Animals were 9-12 months old at the start of the experiment. Mice were chronically prepared with a cranial window through which 2-Dimensional optical imaging spectroscopy (2D-OIS) was used to generate functional maps of the cerebral blood volume and saturation changes evoked by whisker stimulation and vascular reactivity challenges. Unexpectedly, the hemodynamic responses were largely preserved in the J20-AD group. This result failed to confirm previous investigations using the J20-AD model. However, a final acute electrophysiology and 2D-OIS experiment was performed to measure both neural and hemodynamic responses concurrently. In this experiment, previously reported deficits in neurovascular coupling in the J20-AD model were observed. This suggests that J20-AD mice may be more susceptible to the physiologically stressing conditions of an acute experimental procedure compared to WT animals. These results therefore highlight the importance of experimental procedure when determining the characteristics of animal models of human disease.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Circulación Cerebrovascular , Acoplamiento Neurovascular , Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/etiología , Animales , Volumen Sanguíneo Cerebral , Modelos Animales de Enfermedad , Fenómenos Electrofisiológicos , Hemodinámica , Hipercapnia , Masculino , Ratones , Ratones Transgénicos , Método de Montecarlo , Imagen Óptica , Oxígeno/metabolismo , Factores de Tiempo
11.
J Alzheimers Dis ; 68(2): 609-624, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30814360

RESUMEN

The role of cellular changes in the neurovascular unit is increasingly being investigated to understand the pathogenesis of Alzheimer's disease (AD). The aim of the current study was to determine the time course of recognition memory impairment in the J20 mouse model of AD, in relation to neuroinflammatory responses and the pathology of amyloid-ß (Aß). Male hAPP-J20 and wild-type mice were assessed at 3, 6, 9, and 12 months of age. The spontaneous object recognition (SOR) task provided a measure of memory, with assessment of both a short delay (1 min) and a long delay (4 h). Immunohistochemistry was used to characterize Aß deposition, and quantify astrocyte and microglial responses. At all ages tested, J20 mice had impaired long-term, but preserved short-term, recognition memory. Wild-types demonstrated preserved long-term memory up to 9 months of age, and preserved short-term memory at all ages tested. Plaque pathology in the J20 mice was present from 6 months onwards, with co-localization of reactive microglia and activated astrocytes. Reactive microglia and astrocyte activation in the hippocampus were significantly greater in the J20 mice at 9 months, compared to wild-types. This study contributes to our understanding of the pathological and cognitive mechanisms at play in AD. J20 mice showed impairment in retaining information over longer periods from an early age, preceding the deposition of Aß and glial activation. Defining early physiological changes in relation to cognitive decline could provide insight into new therapeutic targets early in the disease process, when intervention is most likely to effectively slow disease progression.


Asunto(s)
Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/psicología , Encéfalo/patología , Neuroglía/patología , Reconocimiento en Psicología , Enfermedad de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Gliosis/metabolismo , Gliosis/patología , Gliosis/psicología , Humanos , Inflamación/metabolismo , Inflamación/patología , Inflamación/psicología , Masculino , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/patología , Trastornos de la Memoria/psicología , Memoria a Largo Plazo , Memoria a Corto Plazo , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuroglía/metabolismo , Factores de Tiempo
12.
BMC Neurosci ; 19(1): 62, 2018 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-30333009

RESUMEN

Efficient blood supply to the brain is of paramount importance to its normal functioning and improper blood flow can result in potentially devastating neurological consequences. Cerebral blood flow in response to neural activity is intrinsically regulated by a complex interplay between various cell types within the brain in a relationship termed neurovascular coupling. The breakdown of neurovascular coupling is evident across a wide variety of both neurological and psychiatric disorders including Alzheimer's disease. Atherosclerosis is a chronic syndrome affecting the integrity and function of major blood vessels including those that supply the brain, and it is therefore hypothesised that atherosclerosis impairs cerebral blood flow and neurovascular coupling leading to cerebrovascular dysfunction. This review will discuss the mechanisms of neurovascular coupling in health and disease and how atherosclerosis can potentially cause cerebrovascular dysfunction that may lead to cognitive decline as well as stroke. Understanding the mechanisms of neurovascular coupling in health and disease may enable us to develop potential therapies to prevent the breakdown of neurovascular coupling in the treatment of vascular brain diseases including vascular dementia, Alzheimer's disease and stroke.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Aterosclerosis/fisiopatología , Demencia Vascular/fisiopatología , Acoplamiento Neurovascular/fisiología , Animales , Encéfalo/irrigación sanguínea , Encéfalo/fisiopatología , Humanos
13.
Front Neurosci ; 12: 550, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30154690

RESUMEN

Anesthetized rodent models are ubiquitous in pre-clinical neuroimaging studies. However, because the associated cerebral morphology and experimental methodology results in a profound negative brain-core temperature differential, cerebral temperature changes during functional activation are likely to be principally driven by local inflow of fresh, core-temperature, blood. This presents a confound to the interpretation of blood-oxygenation level-dependent (BOLD) functional magnetic resonance imaging (fMRI) data acquired from such models, since this signal is also critically temperature-dependent. Nevertheless, previous investigation on the subject is surprisingly sparse. Here, we address this issue through use of a novel multi-modal methodology in the urethane anesthetized rat. We reveal that sensory stimulation, hypercapnia and recurrent acute seizures induce significant increases in cortical temperature that are preferentially correlated to changes in total hemoglobin concentration (Hbt), relative to cerebral blood flow and oxidative metabolism. Furthermore, using a phantom-based evaluation of the effect of such temperature changes on the BOLD fMRI signal, we demonstrate a robust inverse relationship between both variables. These findings suggest that temperature increases, due to functional hyperemia, should be accounted for to ensure accurate interpretation of BOLD fMRI signals in pre-clinical neuroimaging studies.

14.
Clin Sci (Lond) ; 132(8): 851-868, 2018 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-29712883

RESUMEN

Cerebral small vessel disease (SVD) is a major contributor to stroke, cognitive impairment and dementia with limited therapeutic interventions. There is a critical need to provide mechanistic insight and improve translation between pre-clinical research and the clinic. A 2-day workshop was held which brought together experts from several disciplines in cerebrovascular disease, dementia and cardiovascular biology, to highlight current advances in these fields, explore synergies and scope for development. These proceedings provide a summary of key talks at the workshop with a particular focus on animal models of cerebral vascular disease and dementia, mechanisms and approaches to improve translation. The outcomes of discussion groups on related themes to identify the gaps in knowledge and requirements to advance knowledge are summarized.


Asunto(s)
Enfermedades de los Pequeños Vasos Cerebrales/etiología , Investigación Biomédica Traslacional , Animales , Humanos
15.
Neuroimage ; 171: 165-175, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29294386

RESUMEN

Whether functional hyperemia during epileptic activity is adequate to meet the heightened metabolic demand of such events is controversial. Whereas some studies have demonstrated hyperoxia during ictal onsets, other work has reported transient hypoxic episodes that are spatially dependent on local surface microvasculature. Crucially, how laminar differences in ictal evolution can affect subsequent cerebrovascular responses has not been thus far investigated, and is likely significant in view of possible laminar-dependent neurovascular mechanisms and angioarchitecture. We addressed this open question using a novel multi-modal methodology enabling concurrent measurement of cortical tissue oxygenation, blood flow and hemoglobin concentration, alongside laminar recordings of neural activity, in a urethane anesthetized rat model of recurrent seizures induced by 4-aminopyridine. We reveal there to be a close relationship between seizure epicenter depth, translaminar local field potential (LFP) synchrony and tissue oxygenation during the early stages of recurrent seizures, whereby deep layer seizures are associated with decreased cross laminar synchrony and prolonged periods of hypoxia, and middle layer seizures are accompanied by increased cross-laminar synchrony and hyperoxia. Through comparison with functional activation by somatosensory stimulation and graded hypercapnia, we show that these seizure-related cerebrovascular responses occur in the presence of conserved neural-hemodynamic and blood flow-volume coupling. Our data provide new insights into the laminar dependency of seizure-related neurovascular responses, which may reconcile inconsistent observations of seizure-related hypoxia in the literature, and highlight a potential layer-dependent vulnerability that may contribute to the harmful effects of clinical recurrent seizures. The relevance of our findings to perfusion-related functional neuroimaging techniques in epilepsy are also discussed.


Asunto(s)
Encéfalo/irrigación sanguínea , Encéfalo/fisiopatología , Hiperoxia/fisiopatología , Convulsiones/fisiopatología , Animales , Circulación Cerebrovascular/fisiología , Femenino , Hemodinámica/fisiología , Ratas
16.
Neuroimage ; 146: 575-588, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27646129

RESUMEN

It is generally recognised that event related potentials (ERPs) of electroencephalogram (EEG) primarily reflect summed post-synaptic activity of the local pyramidal neural population(s). However, it is still not understood how the positive and negative deflections (e.g. P1, N1 etc) observed in ERP recordings are related to the underlying excitatory and inhibitory post-synaptic activity. We investigated the neurogenesis of P1 and N1 in ERPs by pharmacologically manipulating inhibitory post-synaptic activity in the somatosensory cortex of rodent, and concurrently recording EEG and local field potentials (LFPs). We found that the P1 wave in the ERP and LFP of the supragranular layers is determined solely by the excitatory post-synaptic activity of the local pyramidal neural population, as is the initial segment of the N1 wave across cortical depth. The later part of the N1 wave was modulated by inhibitory post-synaptic activity, with its peak and the pulse width increasing as inhibition was reduced. These findings suggest that the temporal delay of inhibition with respect to excitation observed in intracellular recordings is also reflected in extracellular field potentials (FPs), resulting in a temporal window during which only excitatory post-synaptic activity and leak channel activity are recorded in the ERP and evoked LFP time series. Based on these findings, we provide clarification on the interpretation of P1 and N1 in terms of the excitatory and inhibitory post-synaptic activities of the local pyramidal neural population(s).


Asunto(s)
Ondas Encefálicas , Potenciales Evocados Somatosensoriales , Corteza Somatosensorial/fisiología , Animales , Electroencefalografía , Femenino , Inhibición Neural , Estimulación Física , Ratas , Percepción del Tacto/fisiología
17.
J Neuroinflammation ; 13(1): 195, 2016 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-27557843

RESUMEN

BACKGROUND: Neurovascular coupling describes the mechanism by which the energy and oxygen demand arising from neuronal activity is met by an increase in regional blood flow, known as the haemodynamic response. Interleukin 1 (IL-1) is a pro-inflammatory cytokine and an important mediator of neuronal injury, though mechanisms through which IL-1 exerts its effects in the brain are not fully understood. In this study, we set out to investigate if increased cerebral levels of IL-1 have a negative effect on the neurovascular coupling in the cortex in response to sensory stimulation. METHODS: We used two approaches to measure the neuronal activity and haemodynamic changes in the anaesthetised rat barrel somatosensory cortex in response to mechanical whisker stimulation, before and for 6 h after intra-striatal injection of interleukin-1ß or vehicle. First, we used two-dimensional optical imaging spectroscopy (2D-OIS) to measure the size of the functional haemodynamic response, indicated by changes of oxyhaemoglobin (HbO2) and total haemoglobin (HbT) concentration. In the same animals, immunostaining of immunoglobulin G and SJC-positive extravasated neutrophils was used to confirm the pro-inflammatory effects of interleukin-1ß (IL-1ß). Second, to examine the functional coupling between neuronal activity and the haemodynamic response, we used a 'Clark-style' electrode combined with a single sharp electrode to simultaneously record local tissue oxygenation (partial pressure oxygen, pO2) in layer IV/V of the stimulated barrel cortex and multi-unit activity (MUA) together with local field potentials (LFPs), respectively. RESULTS: 2D-OIS data revealed that the size of the haemodynamic response to mechanical whisker stimulation declined over the 6 h following IL-1ß injection whereas the vehicle group remained stable, significant differences being seen after 5 h. Moreover, the size of the transient increases of neuronal LFP activity in response to whisker stimulation decreased after IL-1ß injection, significant changes compared to vehicle being seen for gamma-band activity after 1 h and beta-band activity after 3 h. The amplitude of the functional pO2 response similarly decreased after 3 h post-IL-1ß injection, whereas IL-1ß had no significant effect on the peak of whisker-stimulation-induced MUA. The stimulation-evoked increases in gamma power and pO2 correlated significantly throughout the 6 h in the vehicle group, but such a correlation was not observed in the IL-1ß-injected group. CONCLUSIONS: We conclude that intra-striatal IL-1ß decouples cortical neuronal activity from its haemodynamic response. This finding may have implications for neurological conditions where IL-1ß plays a part, especially those involving reductions in cerebral blood flow (such as stroke).


Asunto(s)
Cuerpo Estriado/efectos de los fármacos , Ritmo Gamma/efectos de los fármacos , Hemodinámica/efectos de los fármacos , Interleucina-1beta/farmacología , Oxígeno/administración & dosificación , Corteza Somatosensorial/fisiología , Análisis de Varianza , Animales , Circulación Cerebrovascular/fisiología , Ritmo Gamma/fisiología , Hemodinámica/fisiología , Hemoglobinas/metabolismo , Inmunoglobulina G/metabolismo , Masculino , Neutrófilos/metabolismo , Oxihemoglobinas/metabolismo , Estimulación Física , Ratas , Corteza Somatosensorial/efectos de los fármacos , Análisis Espectral , Vibrisas/inervación
19.
Sci Rep ; 5: 11990, 2015 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-26189707

RESUMEN

The blood-brain barrier (BBB) is by far the most important target in developing new approaches to improve delivery of drugs and diagnostic tools into the Central Nervous System (CNS). Here we report the engineering of pH- sensitive polymersomes (synthetic vesicles formed by amphiphilic copolymers) that exploit endogenous transport mechanisms to traverse the BBB, enabling delivery of large macromolecules into both the CNS parenchyma and CNS cells. We achieve this by targeting the Low Density Lipoprotein Receptor-Related Protein 1 (LRP-1) receptor. We show that LRP-1 is associated with endothelial transcytosis that does not involve acidification of cargo in membrane-trafficking organelles. By contrast, this receptor is also associated with traditional endocytosis in CNS cells, thus aiding the delivery of relevant cargo within their cytosol. We prove this using IgG as a model cargo, thus demonstrating that the combination of appropriate targeting combined with pH-sensitive polymersomes enables the efficient delivery of macromolecules into CNS cells.


Asunto(s)
Anticuerpos/administración & dosificación , Anticuerpos/metabolismo , Sistema Nervioso Central/metabolismo , Receptores de LDL/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Transporte Biológico , Barrera Hematoencefálica/metabolismo , Espacio Intracelular/metabolismo , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Proteína 1 de la Membrana Asociada a los Lisosomas/metabolismo , Masculino , Ratones , Polímeros/metabolismo , Unión Proteica , Transporte de Proteínas , Transcitosis , Proteínas de Unión al GTP rab/metabolismo
20.
Sci Rep ; 5: 12621, 2015 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-26218081

RESUMEN

Neural activity is closely followed by a localised change in cerebral blood flow, a process termed neurovascular coupling. These hemodynamic changes form the basis of contrast in functional magnetic resonance imaging (fMRI) and are used as a correlate for neural activity. Anesthesia is widely employed in animal fMRI and neurovascular studies, however anesthetics are known to profoundly affect neural and vascular physiology, particularly in mice. Therefore, we investigated the efficacy of a novel 'modular' anesthesia that combined injectable (fentanyl-fluanisone/midazolam) and volatile (isoflurane) anesthetics in mice. To characterize sensory-evoked cortical hemodynamic responses, we used optical imaging spectroscopy to produce functional maps of changes in tissue oxygenation and blood volume in response to mechanical whisker stimulation. Following fine-tuning of the anesthetic regime, stimulation elicited large and robust hemodynamic responses in the somatosensory cortex, characterized by fast arterial activation, increases in total and oxygenated hemoglobin, and decreases in deoxygenated hemoglobin. Overall, the magnitude and speed of evoked hemodynamic responses under anesthesia resembled those in the awake state, indicating that the novel anesthetic combination significantly minimizes the impact of anesthesia. Our findings have broad implications for both neurovascular research and longitudinal fMRI studies that increasingly require the use of genetically engineered mice.


Asunto(s)
Anestesia/métodos , Hemodinámica/fisiología , Corteza Somatosensorial/fisiología , Vigilia/fisiología , Animales , Butirofenonas/administración & dosificación , Circulación Cerebrovascular/efectos de los fármacos , Circulación Cerebrovascular/fisiología , Femenino , Fentanilo/administración & dosificación , Hemodinámica/efectos de los fármacos , Hemoglobinas/metabolismo , Isoflurano/administración & dosificación , Imagen por Resonancia Magnética , Ratones Endogámicos C57BL , Midazolam/administración & dosificación , Oxígeno/metabolismo , Corteza Somatosensorial/irrigación sanguínea , Factores de Tiempo , Vibrisas/efectos de los fármacos , Vibrisas/inervación , Vibrisas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...