Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 432
Filter
1.
Article in English | MEDLINE | ID: mdl-38717876

ABSTRACT

Neurovascular coupling (NVC) provides important insights into the intricate activity of brain functioning and may aid in the early diagnosis of brain diseases. Emerging evidences have shown that NVC could be assessed by the coupling between electroencephalography (EEG) and functional near-infrared spectroscopy (fNIRS). However, this endeavor presents significant challenges due to the absence of standardized methodologies and reliable techniques for coupling analysis of these two modalities. In this study, we introduced a novel method, i.e., the collaborative multi-output variational Gaussian process convergent cross-mapping (CMVGP-CCM) approach to advance coupling analysis of EEG and fNIRS. To validate the robustness and reliability of the CMVGP-CCM method, we conducted extensive experiments using chaotic time series models with varying noise levels, sequence lengths, and causal driving strengths. In addition, we employed the CMVGP-CCM method to explore the NVC between EEG and fNIRS signals collected from 26 healthy participants using a working memory (WM) task. Results revealed a significant causal effect of EEG signals, particularly the delta, theta, and alpha frequency bands, on the fNIRS signals during WM. This influence was notably observed in the frontal lobe, and its strength exhibited a decline as cognitive demands increased. This study illuminates the complex connections between brain electrical activity and cerebral blood flow, offering new insights into the underlying NVC mechanisms of WM.


Subject(s)
Algorithms , Electroencephalography , Memory, Short-Term , Neurovascular Coupling , Spectroscopy, Near-Infrared , Humans , Electroencephalography/methods , Male , Female , Spectroscopy, Near-Infrared/methods , Adult , Normal Distribution , Neurovascular Coupling/physiology , Young Adult , Memory, Short-Term/physiology , Healthy Volunteers , Reproducibility of Results , Multivariate Analysis , Frontal Lobe/physiology , Frontal Lobe/diagnostic imaging , Brain Mapping/methods , Theta Rhythm/physiology , Brain/physiology , Brain/diagnostic imaging , Brain/blood supply , Nonlinear Dynamics , Delta Rhythm/physiology , Alpha Rhythm/physiology
2.
Sci Rep ; 14(1): 11287, 2024 05 17.
Article in English | MEDLINE | ID: mdl-38760449

ABSTRACT

Spectrum power analysis in the low frequency oscillations (LFO) region of functional near infrared spectroscopy (fNIRS) is a promising method to deliver information about brain activation and therefore might be used for prognostication in patients with disorders of consciousness in the neurocritical care unit alongside with established methods. In this study, we measure the cortical hemodynamic response measured by fNIRS in the LFO region following auditory and somatosensory stimulation in healthy subjects. The significant hemodynamic reaction in the contralateral hemisphere correlation with the physiologic electric response suggests neurovascular coupling. In addition, we investigate power spectrum changes in steady state measurements of cerebral death patients and healthy subjects in the LFO region, the frequency of the heartbeat and respiration. The spectral power within the LFO region was lower in the patients with cerebral death compared to the healthy subjects, whereas there were no differences in spectral power for physiological activities such as heartbeat and respiration rate. This finding indicates the cerebral origin of our low frequency measurements. Therefore, LFO measurements are a potential method to detect brain activation in patients with disorders of consciousness and cerebral death. However, further studies in patients are needed to investigate its potential clinical use.


Subject(s)
Brain Death , Neurovascular Coupling , Spectroscopy, Near-Infrared , Humans , Male , Female , Adult , Neurovascular Coupling/physiology , Spectroscopy, Near-Infrared/methods , Brain Death/physiopathology , Middle Aged , Hemodynamics/physiology , Aged , Brain/physiopathology , Brain/physiology , Brain/diagnostic imaging , Heart Rate/physiology
3.
Nat Commun ; 15(1): 3526, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38664419

ABSTRACT

Large-scale imaging of brain activity with high spatio-temporal resolution is crucial for advancing our understanding of brain function. The existing neuroimaging techniques are largely limited by restricted field of view, slow imaging speed, or otherwise do not have the adequate spatial resolution to capture brain activities on a capillary and cellular level. To address these limitations, we introduce fluorescence localization microscopy aided with sparsely-labeled red blood cells for cortex-wide morphological and functional cerebral angiography with 4.9 µm spatial resolution and 1 s temporal resolution. When combined with fluorescence calcium imaging, the proposed method enables extended recordings of stimulus-evoked neuro-vascular changes in the murine brain while providing simultaneous multiparametric readings of intracellular neuronal activity, blood flow velocity/direction/volume, and vessel diameter. Owing to its simplicity and versatility, the proposed approach will become an invaluable tool for deciphering the regulation of cortical microcirculation and neurovascular coupling in health and disease.


Subject(s)
Erythrocytes , Microscopy, Fluorescence , Animals , Erythrocytes/metabolism , Erythrocytes/cytology , Microscopy, Fluorescence/methods , Mice , Cerebral Cortex/blood supply , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/metabolism , Male , Mice, Inbred C57BL , Cerebral Angiography/methods , Calcium/metabolism , Cerebrovascular Circulation/physiology , Fluorescent Dyes/chemistry , Neurovascular Coupling/physiology , Neurons/metabolism , Neurons/physiology , Microcirculation
4.
eNeuro ; 11(5)2024 May.
Article in English | MEDLINE | ID: mdl-38641415

ABSTRACT

Both the retina and brain exhibit neurovascular coupling, increased blood flow during increased neural activity. In the retina increased blood flow can be evoked by flickering light, but the magnitude of the metabolic change that underlies this is not known. Local changes in oxygen consumption (QO2) are difficult to measure in vivo when both supply and demand are changing. Here we isolated the C57BL/6J mouse retina and supplied it with oxygen from both sides of the tissue. Microelectrode recordings of PO2 were made in darkness and during 20 s of high scotopic flickering light at 1 Hz. Flicker led to a PO2 increase in the outer retina and a decrease in the inner retina, indicating that outer retinal QO2 (QOR) decreased and inner retinal QO2 (QIR) increased. A four-layer oxygen diffusion model was fitted to PO2 values obtained in darkness and at the end of flicker to determine the values of QOR and QIR. QOR in flicker was 76 ± 14% (mean and SD, n = 10) of QOR in darkness. The increase in QIR was smaller, 6.4 ± 5.0%. These metabolic changes are likely smaller than the maximum changes, because with no regeneration of pigment in the isolated retina, we limited the illumination. Further modeling indicated that at high illumination, QIR could increase by up to 45%, which is comparable to the magnitude of flow changes. This suggests that the blood flow increase is at least roughly matched to the increased metabolic demands of activity in the retina.


Subject(s)
Mice, Inbred C57BL , Oxygen Consumption , Oxygen , Photic Stimulation , Retina , Animals , Retina/metabolism , Oxygen Consumption/physiology , Photic Stimulation/methods , Oxygen/metabolism , Oxygen/blood , Mice , Male , Light , Darkness , Neurovascular Coupling/physiology
5.
Neurosci Bull ; 40(5): 621-634, 2024 May.
Article in English | MEDLINE | ID: mdl-38564049

ABSTRACT

Epilepsy is a multifaceted neurological syndrome characterized by recurrent, spontaneous, and synchronous seizures. The pathogenesis of epilepsy, known as epileptogenesis, involves intricate changes in neurons, neuroglia, and endothelium, leading to structural and functional disorders within neurovascular units and culminating in the development of spontaneous epilepsy. Although current research on epilepsy treatments primarily centers around anti-seizure drugs, it is imperative to seek effective interventions capable of disrupting epileptogenesis. To this end, a comprehensive exploration of the changes and the molecular mechanisms underlying epileptogenesis holds the promise of identifying vital biomarkers for accurate diagnosis and potential therapeutic targets. Emphasizing early diagnosis and timely intervention is paramount, as it stands to significantly improve patient prognosis and alleviate the socioeconomic burden. In this review, we highlight the changes and molecular mechanisms of the neurovascular unit in epileptogenesis and provide a theoretical basis for identifying biomarkers and drug targets.


Subject(s)
Epilepsy , Humans , Epilepsy/physiopathology , Animals , Neurovascular Coupling/physiology , Brain/metabolism , Brain/physiopathology , Anticonvulsants/therapeutic use , Neurons/metabolism , Neuroglia/metabolism
6.
Commun Biol ; 7(1): 287, 2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38459113

ABSTRACT

Dynamic changes in astrocyte Ca2+ are recognized as contributors to functional hyperemia, a critical response to increased neuronal activity mediated by a process known as neurovascular coupling (NVC). Although the critical role of glutamatergic signaling in this process has been extensively investigated, the impact of behavioral state, and the release of behavior-associated neurotransmitters, such as norepinephrine and serotonin, on astrocyte Ca2+ dynamics and functional hyperemia have received less attention. We used two-photon imaging of the barrel cortex in awake mice to examine the role of noradrenergic and serotonergic projections in NVC. We found that both neurotransmitters facilitated sensory stimulation-induced increases in astrocyte Ca2+. Interestingly, while ablation of serotonergic neurons reduced sensory stimulation-induced functional hyperemia, ablation of noradrenergic neurons caused both attenuation and potentiation of functional hyperemia. Our study demonstrates that norepinephrine and serotonin are involved in modulating sensory stimulation-induced astrocyte Ca2+ elevations and identifies their differential effects in regulating functional hyperemia.


Subject(s)
Adrenergic Neurons , Hyperemia , Neurovascular Coupling , Mice , Animals , Neurovascular Coupling/physiology , Serotonin , Neurotransmitter Agents , Norepinephrine , Signal Transduction
7.
J Neurosci ; 44(22)2024 May 29.
Article in English | MEDLINE | ID: mdl-38548341

ABSTRACT

The neurovascular unit (NVU) includes multiple different cell types, including neurons, astrocytes, endothelial cells, and pericytes, which respond to insults on very different time or dose scales. We defined differential vulnerability among these cell types, using response to two different insults: oxygen-glucose deprivation (OGD) and thrombin-mediated cytotoxicity. We found that neurons are most vulnerable, followed by endothelial cells and astrocytes. After temporary focal cerebral ischemia in male rats, we found significantly more injured neurons, compared with astrocytes in the ischemic area, consistent with differential vulnerability in vivo. We sought to illustrate different and shared mechanisms across all cell types during response to insult. We found that gene expression profiles in response to OGD differed among the cell types, with a paucity of gene responses shared by all types. All cell types activated genes relating to autophagy, apoptosis, and necroptosis, but the specific genes differed. Astrocytes and endothelial cells also activated pathways connected to DNA repair and antiapoptosis. Taken together, the data support the concept of differential vulnerability in the NVU and suggest that different elements of the unit will evolve from salvageable to irretrievable on different time scales while residing in the same brain region and receiving the same (ischemic) blood flow. Future work will focus on the mechanisms of these differences. These data suggest future stroke therapy development should target different elements of the NVU differently.


Subject(s)
Astrocytes , Endothelial Cells , Neurons , Rats, Sprague-Dawley , Animals , Male , Rats , Astrocytes/metabolism , Astrocytes/pathology , Endothelial Cells/metabolism , Neurons/metabolism , Brain/metabolism , Brain/pathology , Glucose/deficiency , Glucose/metabolism , Brain Ischemia/pathology , Brain Ischemia/metabolism , Brain Ischemia/genetics , Pericytes/metabolism , Pericytes/pathology , Neurovascular Coupling/physiology
8.
Cell Rep ; 43(4): 113970, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38512868

ABSTRACT

To meet the high energy demands of brain function, cerebral blood flow (CBF) parallels changes in neuronal activity by a mechanism known as neurovascular coupling (NVC). However, which neurons play a role in mediating NVC is not well understood. Here, we identify in mice and humans a specific population of cortical GABAergic neurons that co-express neuronal nitric oxide synthase and tachykinin receptor 1 (Tacr1). Through whole-tissue clearing, we demonstrate that Tacr1 neurons extend local and long-range projections across functionally connected cortical areas. We show that whisker stimulation elicited Tacr1 neuron activity in the barrel cortex through feedforward excitatory pathways. Additionally, through optogenetic experiments, we demonstrate that Tacr1 neurons are instrumental in mediating CBF through the relaxation of mural cells in a similar fashion to whisker stimulation. Finally, by electron microscopy, we observe that Tacr1 processes contact astrocytic endfeet. These findings suggest that Tacr1 neurons integrate cortical activity to mediate NVC.


Subject(s)
Neurovascular Coupling , Animals , Mice , Neurovascular Coupling/physiology , Humans , Neurons/metabolism , Neurons/physiology , Vibrissae/physiology , Mice, Inbred C57BL , GABAergic Neurons/metabolism , GABAergic Neurons/physiology , Male , Cerebral Cortex/physiology , Cerebral Cortex/blood supply , Cerebrovascular Circulation/physiology , Nitric Oxide Synthase Type I/metabolism
9.
CNS Neurosci Ther ; 30(2): e14584, 2024 02.
Article in English | MEDLINE | ID: mdl-38421125

ABSTRACT

AIMS: Most studies focus on dynamic cerebral autoregulation (dCA) in the middle cerebral artery (MCA), and few studies investigated neurovascular coupling (NVC) and dCA in the posterior cerebral artery (PCA). We investigated NVC and dCA of the PCA in healthy volunteers to identify sex differences. METHODS: Thirty men and 30 age-matched women completed dCA and NCV assessments. The cerebral blood flow velocity (CBFV) and mean arterial pressure were evaluated using transcranial Doppler ultrasound and a servo-controlled plethysmograph, respectively. The dCA parameters were analyzed using transfer function analysis. The NCV was evaluated by eyes-open and eyes-closed (24 s each) periodically based on voice prompts. The eyes-open visual stimulation comprised silent reading of Beijing-related tourist information. RESULTS: The PCA gain was lower than that of the MCA in all frequency ranges (all p < 0.05). Phase was consistent across the cerebrovascular territories. The cerebrovascular conductance index (CVCi) and mean CBFV (MV) of the PCA were significantly higher during the eyes-open than eyes-closed period (CVCi: 0.50 ± 0.12 vs. 0.38 ± 0.10; MV: 42.89 ± 8.49 vs. 32.98 ± 7.25, both p < 0.001). The PCA dCA and NVC were similar between the sexes. CONCLUSION: We assessed two major mechanisms that maintain cerebral hemodynamic stability in healthy men and women. The visual stimulation-evoked CBFV of the PCA was significantly increased compared to that during rest, confirming the activation of NVC. Men and women have similar functions in PCA dCA and NCV.


Subject(s)
Neurovascular Coupling , Humans , Male , Female , Neurovascular Coupling/physiology , Posterior Cerebral Artery/diagnostic imaging , Blood Flow Velocity/physiology , Homeostasis/physiology , Middle Cerebral Artery/diagnostic imaging , Middle Cerebral Artery/physiology , Ultrasonography, Doppler, Transcranial , Cerebrovascular Circulation/physiology , Blood Pressure/physiology
10.
Ultrasound Med Biol ; 50(5): 751-759, 2024 05.
Article in English | MEDLINE | ID: mdl-38418342

ABSTRACT

OBJECTIVE: Neurovascular coupling (NVC) represents the increase in regional blood flow associated with neural activity. The aim here was to describe a new approach to non-invasive measurement of NVC by spectral analysis of the cerebral blood flow velocity (CBFV) with transcranial Doppler. METHODS: In a sample of 20 healthy participants, we monitored systolic CBFV in the left posterior cerebral artery (PCA) during off (eyes closed) and on (flickering checkerboard) periods. The contralateral middle cerebral artery was simultaneously monitored as a control. Each participant was submitted to three experiments, each having five cycles, with increasing duration of the cycles, from 10 s (0.1 Hz) to 20 s (0.05 Hz) and lastly 40 s (0.025 Hz), half the time for on and for off periods, constituting a total of 6 min. The successive cycles were expected to cause oscillation in CBFV in a sinusoidal pattern that could be characterized by spectral analysis. We also measured the classic CBFV overshoot as the relative increase in percentage of systolic CBFV from baseline. The relationship and agreement between the two methods were analyzed by linear regression and Bland-Altman plots. In every participant, a clear peak of amplitude in the PCA CBFV spectrum was discernible at 0.1, 0.05 and 0.025 Hz of visual stimulation. RESULTS: On average, this amplitude was 7.1 ± 2.3%, 10.9 ± 3.5% and 17.3 ± 6.5%, respectively. This response contrasted significantly with an absent peak in middle cerebral artery monitoring (p < 0.0001). The spectral amplitude and classic overshoot were highly correlated and linearly related (p < 0.0001). CONCLUSION: NVC can be quantified by the spectral amplitude of PCA CBFV at slower and higher frequencies of visual stimulation. This method represents an alternative to classic overshoot without the need for stimulus marking or synchronization.


Subject(s)
Neurovascular Coupling , Humans , Neurovascular Coupling/physiology , Middle Cerebral Artery/diagnostic imaging , Blood Flow Velocity/physiology , Ultrasonography, Doppler, Transcranial/methods , Healthy Volunteers , Cerebrovascular Circulation/physiology
11.
Biochem Soc Trans ; 52(1): 279-289, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38385536

ABSTRACT

Diet is currently recognized as a major modifiable agent of human health. In particular, dietary nitrate has been increasingly explored as a strategy to modulate different physiological mechanisms with demonstrated benefits in multiple organs, including gastrointestinal, cardiovascular, metabolic, and endocrine systems. An intriguing exception in this scenario has been the brain, for which the evidence of the nitrate benefits remains controversial. Upon consumption, nitrate can undergo sequential reduction reactions in vivo to produce nitric oxide (•NO), a ubiquitous paracrine messenger that supports multiple physiological events such as vasodilation and neuromodulation. In the brain, •NO plays a key role in neurovascular coupling, a fine process associated with the dynamic regulation of cerebral blood flow matching the metabolic needs of neurons and crucial for sustaining brain function. Neurovascular coupling dysregulation has been associated with neurodegeneration and cognitive dysfunction during different pathological conditions and aging. We discuss the potential biological action of nitrate on brain health, concerning the molecular mechanisms underpinning this association, particularly via modulation of •NO-dependent neurovascular coupling. The impact of nitrate supplementation on cognitive performance was scrutinized through preclinical and clinical data, suggesting that intervention length and the health condition of the participants are determinants of the outcome. Also, it stresses the need for multimodal quantitative studies relating cellular and mechanistic approaches to function coupled with behavior clinical outputs to understand whether a mechanistic relationship between dietary nitrate and cognitive health is operative in the brain. If proven, it supports the exciting hypothesis of cognitive enhancement via diet.


Subject(s)
Neurovascular Coupling , Humans , Neurovascular Coupling/physiology , Nitrates/pharmacology , Nitric Oxide/metabolism , Dietary Supplements , Cognition
12.
Neuroimage ; 289: 120535, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38342188

ABSTRACT

Neurovascular coupling serves as an essential neurophysiological mechanism in functional neuroimaging, which is generally presumed to be robust and invariant across different physiological states, encompassing both task engagement and resting state. Nevertheless, emerging evidence suggests that neurovascular coupling may exhibit state dependency, even in normal human participants. To investigate this premise, we analyzed the cross-frequency spectral correspondence between concurrently recorded electroencephalography (EEG) and functional magnetic resonance imaging (fMRI) data, utilizing them as proxies for neurovascular coupling during the two conditions: an eye-open-eye-close (EOEC) task and a resting state. We hypothesized that given the state dependency of neurovascular coupling, EEG-fMRI spectral correspondences would change between the two conditions in the visual system. During the EOEC task, we observed a negative phase-amplitude-coupling (PAC) between EEG alpha-band and fMRI visual activity. Conversely, in the resting state, a pronounced amplitude-amplitude-coupling (AAC) emerged between EEG and fMRI signals, as evidenced by the spectral correspondence between the EEG gamma-band of the midline occipital channel (Oz) and the high-frequency fMRI signals (0.15-0.25 Hz) in the visual network. This study reveals distinct scenarios of EEG-fMRI spectral correspondence in healthy participants, corroborating the state-dependent nature of neurovascular coupling.


Subject(s)
Magnetic Resonance Imaging , Neurovascular Coupling , Humans , Magnetic Resonance Imaging/methods , Neurovascular Coupling/physiology , Brain Mapping/methods , Electroencephalography/methods , Eye , Brain/diagnostic imaging , Brain/physiology
13.
J Cereb Blood Flow Metab ; 44(6): 911-924, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38230631

ABSTRACT

Imaging hemodynamic responses to interictal spikes holds promise for presurgical epilepsy evaluations. Understanding the hemodynamic response function is crucial for accurate interpretation. Prior interictal neurovascular coupling data primarily come from anesthetized animals, impacting reliability. We simultaneously monitored calcium fluctuations in excitatory neurons, hemodynamics, and local field potentials (LFP) during bicuculline-induced interictal events in both isoflurane-anesthetized and awake mice. Isoflurane significantly affected LFP amplitude but had little impact on the amplitude and area of the calcium signal. Anesthesia also dramatically blunted the amplitude and latency of the hemodynamic response, although not its area of spread. Cerebral blood volume change provided the best spatial estimation of excitatory neuronal activity in both states. Targeted silencing of the thalamus in awake mice failed to recapitulate the impact of anesthesia on hemodynamic responses suggesting that isoflurane's interruption of the thalamocortical loop did not contribute either to the dissociation between the LFP and the calcium signal nor to the alterations in interictal neurovascular coupling. The blood volume increase associated with interictal spikes represents a promising mapping signal in both the awake and anesthetized states.


Subject(s)
Hemodynamics , Isoflurane , Neurons , Wakefulness , Animals , Mice , Wakefulness/drug effects , Wakefulness/physiology , Hemodynamics/drug effects , Neurons/drug effects , Isoflurane/pharmacology , Anesthesia , Male , Cerebrovascular Circulation/drug effects , Cerebrovascular Circulation/physiology , Mice, Inbred C57BL , Bicuculline/pharmacology , Neurovascular Coupling/drug effects , Neurovascular Coupling/physiology
14.
Mol Psychiatry ; 29(3): 820-834, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38238549

ABSTRACT

Cocaine affects both cerebral blood vessels and neuronal activity in brain. Cocaine can also disrupt astrocytes, which modulate neurovascular coupling-a process that regulates cerebral hemodynamics in response to neuronal activation. However, separating neuronal and astrocytic effects from cocaine's direct vasoactive effects has been challenging, partially due to limitations of neuroimaging techniques able to differentiate vascular from neuronal and glial effects at high temporal and spatial resolutions. Here, we used a newly-developed multi-channel fluorescence and optical coherence Doppler microscope (fl-ODM) that allows for simultaneous measurements of neuronal and astrocytic activities (reflected by the intracellular calcium changes in neurons Ca2+N and astrocytes Ca2+A, respectively) alongside their vascular interactions in vivo to address this challenge. Using green and red genetically-encoded Ca2+ indicators differentially expressed in astrocytes and neurons, fl-ODM enabled concomitant imaging of large-scale astrocytic and neuronal Ca2+ fluorescence and 3D cerebral blood flow velocity (CBFv) in vascular networks in the mouse cortex. We assessed cocaine's effects in the prefrontal cortex (PFC) and found that the CBFv changes triggered by cocaine were temporally correlated with astrocytic Ca2+A activity. Chemogenetic inhibition of astrocytes during the baseline state resulted in blood vessel dilation and CBFv increases but did not affect neuronal activity, suggesting modulation of spontaneous blood vessel's vascular tone by astrocytes. Chemogenetic inhibition of astrocytes during a cocaine challenge prevented its vasoconstricting effects alongside the CBFv decreases, but it also attenuated the neuronal Ca2+N increases triggered by cocaine. These results document a role of astrocytes both in regulating vascular tone and consequently blood flow, at baseline and for modulating the vasoconstricting and neuronal activation responses to cocaine in the PFC. Strategies to inhibit astrocytic activity could offer promise for ameliorating vascular and neuronal toxicity from cocaine misuse.


Subject(s)
Astrocytes , Calcium , Cerebrovascular Circulation , Cocaine , Neurons , Prefrontal Cortex , Astrocytes/drug effects , Astrocytes/metabolism , Animals , Cocaine/pharmacology , Cerebrovascular Circulation/drug effects , Cerebrovascular Circulation/physiology , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Mice , Neurons/drug effects , Neurons/metabolism , Male , Calcium/metabolism , Mice, Inbred C57BL , Neurovascular Coupling/drug effects , Neurovascular Coupling/physiology
15.
Article in English | MEDLINE | ID: mdl-38231809

ABSTRACT

Neurovascular coupling (NVC) connects neural activity with hemodynamics and plays a vital role in sustaining brain function. Combining electroencephalography (EEG) and functional near-infrared spectroscopy (fNIRS) is a promising way to explore the NVC. However, the high-order property of EEG data and variability of hemodynamic response function (HRF) across subjects have not been well considered in existing NVC studies. In this study, we proposed a novel framework to enhance the subject-specific parametric modeling of NVC from simultaneous EEG-fNIRS measurement. Specifically, task-related tensor decomposition of high-order EEG data was performed to extract the underlying connections in the temporal-spectral-spatial structures of EEG activities and identify the most relevant temporal signature within multiple trials. Subject-specific HRFs were estimated by parameters optimization of a double gamma function model. A canonical motor task experiment was designed to induce neural activity and validate the effectiveness of the proposed framework. The results indicated that the proposed framework significantly improves the reproducibility of EEG components and the correlation between the predicted hemodynamic activities and the real fNIRS signal. Moreover, the estimated parameters characterized the NVC differences in the task with two speeds. Therefore, the proposed framework provides a feasible solution for the quantitative assessment of the NVC function.


Subject(s)
Neurovascular Coupling , Humans , Neurovascular Coupling/physiology , Reproducibility of Results , Spectroscopy, Near-Infrared/methods , Electroencephalography/methods , Hemodynamics/physiology
16.
Nat Neurosci ; 27(2): 232-248, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38168932

ABSTRACT

Neurovascular coupling (NVC) is important for brain function and its dysfunction underlies many neuropathologies. Although cell-type specificity has been implicated in NVC, how active neural information is conveyed to the targeted arterioles in the brain remains poorly understood. Here, using two-photon focal optogenetics in the mouse cerebral cortex, we demonstrate that single glutamatergic axons dilate their innervating arterioles via synaptic-like transmission between neural-arteriolar smooth muscle cell junctions (NsMJs). The presynaptic parental-daughter bouton makes dual innervations on postsynaptic dendrites and on arteriolar smooth muscle cells (aSMCs), which express many types of neuromediator receptors, including a low level of glutamate NMDA receptor subunit 1 (Grin1). Disruption of NsMJ transmission by aSMC-specific knockout of GluN1 diminished optogenetic and whisker stimulation-caused functional hyperemia. Notably, the absence of GluN1 subunit in aSMCs reduced brain atrophy following cerebral ischemia by preventing Ca2+ overload in aSMCs during arteriolar constriction caused by the ischemia-induced spreading depolarization. Our findings reveal that NsMJ transmission drives NVC and open up a new avenue for studying stroke.


Subject(s)
Neurovascular Coupling , Mice , Animals , Neurovascular Coupling/physiology , Vasodilation/physiology , Axons , Synaptic Transmission , Arterioles/metabolism , Myocytes, Smooth Muscle
17.
J Cereb Blood Flow Metab ; 44(2): 155-168, 2024 02.
Article in English | MEDLINE | ID: mdl-37728791

ABSTRACT

Hypoglycemia is a serious complication of insulin treatment of diabetes that can lead to coma and death. Neurovascular coupling, which mediates increased local blood flow in response to neuronal activity, increases glucose availability to active neurons. This mechanism could be essential for neuronal health during hypoglycemia, when total glucose supplies are low. Previous studies suggest, however, that neurovascular coupling (a transient blood flow increase in response to an increase in neuronal activity) may be reduced during hypoglycemia. Such a reduction in blood flow increase would exacerbate the effects of hypoglycemia, depriving active neurons of glucose. We have reexamined the effects of hypoglycemia on neurovascular coupling by simultaneously monitoring neuronal and vascular responses to whisker stimulation in the awake mouse somatosensory cortex. We find that neurovascular coupling at both penetrating arterioles and at 2nd order capillaries did not change significantly during insulin-induced hypoglycemia compared to euglycemia. In addition, we show that the basal diameter of both arterioles and capillaries increases during hypoglycemia (10.3 and 9.7% increases, respectively). Our results demonstrate that both neurovascular coupling and basal increases in vessel diameter are active mechanisms which help to maintain an adequate supply of glucose to the brain during hypoglycemia.


Subject(s)
Hypoglycemia , Insulins , Neurovascular Coupling , Mice , Animals , Neurovascular Coupling/physiology , Arterioles/metabolism , Capillaries/metabolism , Cerebrovascular Circulation/physiology , Vibrissae/physiology , Hypoglycemia/chemically induced , Hypoglycemia/metabolism , Glucose/metabolism , Insulins/metabolism , Insulins/pharmacology
18.
Adv Sci (Weinh) ; 11(10): e2303516, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38155460

ABSTRACT

Impaired cerebrovascular function contributes to the genesis of age-related cognitive decline. In this study, the hypothesis is tested that impairments in neurovascular coupling (NVC) responses and brain network function predict cognitive dysfunction in older adults. Cerebromicrovascular and working memory function of healthy young (n = 21, 33.2±7.0 years) and aged (n = 30, 75.9±6.9 years) participants are assessed. To determine NVC responses and functional connectivity (FC) during a working memory (n-back) paradigm, oxy- and deoxyhemoglobin concentration changes from the frontal cortex using functional near-infrared spectroscopy are recorded. NVC responses are significantly impaired during the 2-back task in aged participants, while the frontal networks are characterized by higher local and global connection strength, and dynamic FC (p < 0.05). Both impaired NVC and increased FC correlate with age-related decline in accuracy during the 2-back task. These findings suggest that task-related brain states in older adults require stronger functional connections to compensate for the attenuated NVC responses associated with working memory load.


Subject(s)
Cognitive Dysfunction , Neurovascular Coupling , Humans , Aged , Neurovascular Coupling/physiology , Brain/physiology , Frontal Lobe
19.
Geroscience ; 46(1): 327-347, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38123890

ABSTRACT

Age-related impairment of neurovascular coupling (NVC; "functional hyperemia") is a critical factor in the development of vascular cognitive impairment (VCI). Recent geroscience research indicates that cell-autonomous mechanisms alone cannot explain all aspects of neurovascular aging. Circulating factors derived from other organs, including pro-geronic factors (increased with age and detrimental to vascular homeostasis) and anti-geronic factors (preventing cellular aging phenotypes and declining with age), are thought to orchestrate cellular aging processes. This study aimed to investigate the influence of age-related changes in circulating factors on neurovascular aging. Heterochronic parabiosis was utilized to assess how exposure to young or old systemic environments could modulate neurovascular aging. Results demonstrated a significant decline in NVC responses in aged mice subjected to isochronic parabiosis (20-month-old C57BL/6 mice [A-(A)]; 6 weeks of parabiosis) when compared to young isochronic parabionts (6-month-old, [Y-(Y)]). However, exposure to young blood from parabionts significantly improved NVC in aged heterochronic parabionts [A-(Y)]. Conversely, young mice exposed to old blood from aged parabionts exhibited impaired NVC responses [Y-(A)]. In conclusion, even a brief exposure to a youthful humoral environment can mitigate neurovascular aging phenotypes, rejuvenating NVC responses. Conversely, short-term exposure to an aged humoral milieu in young mice accelerates the acquisition of neurovascular aging traits. These findings highlight the plasticity of neurovascular aging and suggest the presence of circulating anti-geronic factors capable of rejuvenating the aging cerebral microcirculation. Further research is needed to explore whether young blood factors can extend their rejuvenating effects to address other age-related cerebromicrovascular pathologies, such as blood-brain barrier integrity.


Subject(s)
Neurovascular Coupling , Mice , Animals , Neurovascular Coupling/physiology , Rejuvenation , Mice, Inbred C57BL , Aging/physiology , Parabiosis
20.
Sheng Li Xue Bao ; 75(6): 903-917, 2023 Dec 25.
Article in Chinese | MEDLINE | ID: mdl-38151352

ABSTRACT

Aging is a natural process accompanied with a progressive deterioration of cognitive functions. With an aging population, more and more elderly people are suffering from cognitive impairment. Previous studies have paid more attention to the impact of inflammation and oxidative stress on cognitive function during aging. Recently, it has been discovered that neurovascular coupling (NVC), a mechanism regulating cerebral blood flow, may play a significant role in aging-related cognitive impairment. NVC responses regulate the supply of energy substances and oxygen during brain activity, which in turn enhances cognitive function. However, as people grow older, NVC responses gradually weaken, which may be one of the mechanisms underlying aging-induced cognitive impairment. Given the important role of NVC responses in the brain, it is necessary to search for intervention methods that can improve NVC responses and promote cognitive function. Exercise is an effective means to delay aging and improve cognitive function. It also has a certain promoting effect on NVC responses. This article reviews the regulatory mechanisms of NVC responses, the relationship between NVC responses and cognitive function, and explores the effects of aging and exercise intervention on NVC responses, hoping to provide new research ideas for exercise intervention to improve NVC responses and promote cognitive function in the elderly.


Subject(s)
Neurovascular Coupling , Humans , Aged , Neurovascular Coupling/physiology , Aging , Cerebrovascular Circulation/physiology , Cognition , Brain
SELECTION OF CITATIONS
SEARCH DETAIL
...