Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
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
2.
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
3.
Int J Mol Sci ; 23(3)2022 Jan 25.
Article in English | MEDLINE | ID: mdl-35163285

ABSTRACT

We investigated the effect of tofogliflozin, a sodium-dependent glucose cotransporter 2 inhibitor (SGLT2i), on retinal blood flow dysregulation, neural retinal dysfunction, and the impaired neurovascular coupling in type 2 diabetic mice. Tofogliflozin was added to mouse chow to deliver 5 mg/kg/day and 6-week-old mice were fed for 8 weeks. The longitudinal changes in the retinal neuronal function and blood flow responses to systemic hyperoxia and flicker stimulation were evaluated every 2 weeks in diabetic db/db mice that received tofogliflozin (n =6) or placebo (n = 6) from 8 to 14 weeks of age. We also evaluated glial activation and vascular endothelial growth factor (VEGF) expression by immunofluorescence. Tofogliflozin treatment caused a sustained decrease in blood glucose in db/db mice from 8 weeks of the treatment. In tofogliflozin-treated db/db mice, both responses improved from 8 to 14 weeks of age, compared with vehicle-treated diabetic mice. Subsequently, the electroretinography implicit time for the oscillatory potential was significantly improved in SGLT2i-treated db/db mice. The systemic tofogliflozin treatment prevented the activation of glial fibrillary acidic protein and VEGF protein expression, as detected by immunofluorescence. Our results suggest that glycemic control with tofogliflozin significantly improved the impaired retinal neurovascular coupling in type 2 diabetic mice with the inhibition of retinal glial activation.


Subject(s)
Benzhydryl Compounds/pharmacology , Glucosides/pharmacology , Neurovascular Coupling/physiology , Sodium-Glucose Transporter 2/metabolism , Animals , Benzhydryl Compounds/metabolism , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Diabetic Retinopathy/prevention & control , Glucosides/metabolism , Hypoglycemic Agents/pharmacology , Male , Mice , Mice, Inbred C57BL , Neurovascular Coupling/drug effects , Retina/drug effects , Retina/metabolism , Sodium-Glucose Transport Proteins/antagonists & inhibitors , Sodium-Glucose Transport Proteins/metabolism , Sodium-Glucose Transporter 2/drug effects , Sodium-Glucose Transporter 2 Inhibitors/metabolism , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Vascular Endothelial Growth Factor A/drug effects , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
4.
Neuroimage ; 245: 118769, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34861394

ABSTRACT

The effects of hippocampal neuronal afterdischarges (nAD) on hemodynamic parameters, such as blood-oxygen-level-dependent (BOLD) signals) and local cerebral blood volume (CBV) changes, as well as neuronal activity and metabolic parameters in the dentate gyrus, was investigated in rats by combining in vivo electrophysiology with functional magnetic resonance imaging (fMRI) or 1H-nuclear magnetic resonance spectroscopy (1H-NMRS). Brief electrical high-frequency pulse-burst stimulation of the right perforant pathway triggered nAD, a seizure-like activity, in the right dentate gyrus with a high incidence, a phenomenon that in turn caused a sustained decrease in BOLD signals for more than 30 min. The decrease was associated with a reduction in CBV but not with signs of hypoxic metabolism. nAD also triggered transient changes mainly in the low gamma frequency band that recovered within 20 min, so that the longer-lasting altered hemodynamics reflected a switch in blood supply rather than transient changes in ongoing neuronal activity. Even in the presence of reduced baseline BOLD signals, neurovascular coupling mechanisms remained intact, making long-lasting vasospasm unlikely. Subsequently generated nAD did not further alter the baseline BOLD signals. Similarly, nAD did not alter baseline BOLD signals when acetaminophen was previously administered, because acetaminophen alone had already caused a similar decrease in baseline BOLD signals as observed after the first nAD. Thus, at least two different blood supply states exist for the hippocampus, one low and one high, with both states allowing similar neuronal activity. Both acetaminophen and nAD switch from the high to the low blood supply state. As a result, the hemodynamic response function to an identical stimulus differed after nAD or acetaminophen, although the triggered neuronal activity was similar.


Subject(s)
Brain Waves/physiology , Electrocorticography , Hippocampus/physiology , Magnetic Resonance Imaging , Neuroimaging , Neurovascular Coupling/physiology , Proton Magnetic Resonance Spectroscopy , Seizures/physiopathology , Animals , Brain Waves/drug effects , Disease Models, Animal , Hippocampus/drug effects , Male , Neurovascular Coupling/drug effects , Rats , Rats, Wistar , Seizures/metabolism
5.
Eur J Pharmacol ; 910: 174483, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34481878

ABSTRACT

The concept of "Neurovascular Unit" (NVU) was put forward, so that the research goal of Central Nervous System (CNS) diseases gradually transitioned from a single neuron to the structural and functional integrity of the NVU. Zebrafish has the advantages of high homology with human genes, strong reproductive capacity and visualization of neural circuits, so it has become an emerging model organism for NVU research and has been applied to a variety of CNS diseases. Based on CNKI (https://www.cnki.net/) and PubMed (https://pubmed.ncbi.nlm.nih.gov/about/) databases, the author of this article sorted out the relevant literature, analyzed the construction of a zebrafish model of various CNS diseases,and the use of diagrams showed the application of zebrafish in the NVU, revealed its relationship, which would provide new methods and references for the treatment and research of CNS diseases.


Subject(s)
Central Nervous System Agents/pharmacology , Central Nervous System Diseases/physiopathology , Central Nervous System/physiology , Neurovascular Coupling/physiology , Zebrafish/physiology , Animals , Blood-Brain Barrier/cytology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiology , Central Nervous System/blood supply , Central Nervous System/drug effects , Central Nervous System Agents/therapeutic use , Central Nervous System Diseases/drug therapy , Disease Models, Animal , Humans , Microglia/drug effects , Microglia/physiology , Models, Animal , Neurons/drug effects , Neurons/physiology , Neurovascular Coupling/drug effects
6.
Am J Physiol Regul Integr Comp Physiol ; 321(2): R208-R219, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34161746

ABSTRACT

Structural and functional changes in the cerebral vasculature occur with advancing age, which may lead to impaired neurovascular coupling (NVC) and cognitive decline. Cyclooxygenase (COX) inhibition abolishes age-related differences in cerebrovascular reactivity, but it is unclear if COX inhibition impacts NVC. The purpose of this study was to examine the influence of aging on NVC before and after COX inhibition. Twenty-three young (age = 25 ± 4 yr) and 21 older (age = 64 ± 5 yr) adults completed two levels of difficulty of the Stroop and n-back tests before and after COX inhibition. Middle cerebral artery blood velocity (MCAv) was measured using transcranial Doppler ultrasound and mean arterial blood pressure (MAP) was measured using a finger cuff. Hemodynamic variables were measured at rest and in response to cognitive challenges. During the Stroop test, older adults demonstrated a greater increase in MCAv (young: 2.2 ± 6.8% vs. older: 5.9 ± 5.8%; P = 0.030) and MAP (young: 2.0 ± 4.9% vs. older: 4.8 ± 4.9%; P = 0.036) compared with young adults. There were no age-related differences during the n-back test. COX inhibition reduced MCAv by 30% in young and 26% in older adults (P < 0.001 for both). During COX inhibition, there were no age-related differences in the percent change in MCAv or MAP in response to the cognitive tests. Our results show that older adults require greater increases in MCAv and MAP during a test of executive function compared with young adults and that any age-related differences in NVC were abolished during COX inhibition. Collectively, this suggests that aging is associated with greater NVC necessary to accomplish a cognitive task.


Subject(s)
Cerebrovascular Circulation/drug effects , Cognition , Cognitive Aging/psychology , Cyclooxygenase Inhibitors/pharmacology , Hemodynamics/drug effects , Indomethacin/pharmacology , Middle Cerebral Artery/drug effects , Neurovascular Coupling/drug effects , Adolescent , Adult , Age Factors , Aged , Executive Function , Female , Humans , Male , Memory, Short-Term , Middle Aged , Middle Cerebral Artery/diagnostic imaging , Stroop Test , Time Factors , Young Adult
7.
J Neurosci ; 41(19): 4305-4320, 2021 05 12.
Article in English | MEDLINE | ID: mdl-33888602

ABSTRACT

Vascular dysfunction is a universal feature of aging and decreased cerebral blood flow has been identified as an early event in the pathogenesis of Alzheimer's disease (AD). Cerebrovascular dysfunction in AD includes deficits in neurovascular coupling (NVC), a mechanism that ensures rapid delivery of energy substrates to active neurons through the blood supply. The mechanisms underlying NVC impairment in AD, however, are not well understood. We have previously shown that mechanistic/mammalian target of rapamycin (mTOR) drives cerebrovascular dysfunction in models of AD by reducing the activity of endothelial nitric oxide synthase (eNOS), and that attenuation of mTOR activity with rapamycin is sufficient to restore eNOS-dependent cerebrovascular function. Here we show mTOR drives NVC impairments in an AD model through the inhibition of neuronal NOS (nNOS)- and non-NOS-dependent components of NVC, and that mTOR attenuation with rapamycin is sufficient to restore NVC and even enhance it above WT responses. Restoration of NVC and concomitant reduction of cortical amyloid-ß levels effectively treated memory deficits in 12-month-old hAPP(J20) mice. These data indicate that mTOR is a critical driver of NVC dysfunction and underlies cognitive impairment in an AD model. Together with our previous findings, the present studies suggest that mTOR promotes cerebrovascular dysfunction in AD, which is associated with early disruption of nNOS activation, through its broad negative impact on nNOS as well as on non-NOS components of NVC. Our studies highlight the potential of mTOR attenuation as an efficacious treatment for AD and potentially other neurologic diseases of aging.SIGNIFICANCE STATEMENT Failure of the blood flow response to neuronal activation [neurovascular coupling (NVC)] in a model of AD precedes the onset of AD-like cognitive symptoms and is driven, to a large extent, by mammalian/mechanistic target of rapamycin (mTOR)-dependent inhibition of nitric oxide synthase activity. Our studies show that mTOR also drives AD-like failure of non-nitric oxide (NO)-mediated components of NVC. Thus, mTOR attenuation may serve to treat AD, where we find that neuronal NO synthase is profoundly reduced early in disease progression, and potentially other neurologic diseases of aging with cerebrovascular dysfunction as part of their etiology.


Subject(s)
Alzheimer Disease/drug therapy , Memory Disorders/drug therapy , Neurovascular Coupling/drug effects , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Aged , Aged, 80 and over , Alzheimer Disease/psychology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Cerebrovascular Disorders/physiopathology , Cognitive Dysfunction/genetics , Cognitive Dysfunction/psychology , Fear/psychology , Female , Humans , Male , Memory Disorders/psychology , Mice , Mice, Transgenic , Microvessels/pathology , Microvessels/ultrastructure , Nitric Oxide Synthase Type III/metabolism , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases/genetics
8.
Exp Neurol ; 339: 113634, 2021 05.
Article in English | MEDLINE | ID: mdl-33549548

ABSTRACT

Migraine is associated with the activation and sensitisation of the trigeminovascular system and is often accompanied by mechanical hyperalgesia and allodynia. The mechanisms of mechanotransduction during a migraine attack are yet unknown. We have proposed that the ion channel Piezo1 may be involved, since it is expressed in endothelial cells as well as in trigeminal ganglion neurons, and thus, may contribute to the activation of both the vascular and neuronal component of the trigeminovascular system. We took advantage of extracellular recordings from the trigeminocervical complex - a key relay centre in the migraine pain pathway, to directly assess the impact of the differently applied Piezo1 agonist Yoda1 on the sensory processing at the spinal level. At a low dose, Yoda1 slightly facilitated the ongoing firing of central trigeminovascular neurons, however, at a high dose, this substance contributed to the suppression of their activity. Using intravital microscopy, we have revealed that Yoda1 at high dose can also induce the dilation of meningeal arteries innervated by trigeminal afferents. Collectively, here we have identified both neuronal and vascular modulation via selective activation of mechanosensitive Piezo1 channels, which provide new evidence in favour of the Piezo1 role in migraine pathogenesis. We propose several mechanisms that may underlie the revealed effects of Yoda1.


Subject(s)
Intravital Microscopy/methods , Membrane Proteins/agonists , Mesenteric Arteries/drug effects , Neurovascular Coupling/drug effects , Pyrazines/pharmacology , Thiadiazoles/pharmacology , Trigeminal Ganglion/drug effects , Animals , Electrophysiological Phenomena/drug effects , Electrophysiological Phenomena/physiology , Male , Membrane Proteins/physiology , Mesenteric Arteries/physiology , Neurovascular Coupling/physiology , Rats , Rats, Wistar , Trigeminal Ganglion/physiology
9.
CNS Neurosci Ther ; 27(1): 134-144, 2021 01.
Article in English | MEDLINE | ID: mdl-33421349

ABSTRACT

INTRODUCTION: Perampanel is a highly selective and noncompetitive α-amino-3 -hydroxy-5-methyl-4-isoxazole propionate receptor (AMPAR) antagonist, which has been used as an orally administered antiepileptic drug in more than 55 countries. Recently, perampanel was shown to exert neuroprotective effects in hemorrhagic and ischemic stroke models via regulating blood-brain barrier (BBB) function. AIM: Here, the protective effects of perampanel were investigated in an in vitro neurovascular unit (NVU) system established using a triple cell co-culture model (neurons, astrocytes, and brain microvascular endothelial cells) and in an in vivo traumatic brain injury (TBI) model. RESULTS: Neurons in the NVU system exhibit a more mature morphological phenotype compared with neurons cultured alone, and the co-culture system mimicked an impermeable barrier in vitro. Perampanel protects the NVU system against traumatic and excitotoxic injury, as evidenced by reduced lactate dehydrogenase (LDH) release and apoptotic rate. Treatment with perampanel attenuated lipid peroxidation and expression of inflammatory cytokines. In addition, perampanel increased Sirt3 protein expression, enhanced the activities of mitochondrial enzyme IDH2 and SOD2, and preserved BBB function in vitro. Knockdown of Sirt3 using specific siRNA (Si-Sirt3) partially reserved the effects of perampanel on neuronal injury and BBB function. Treatment with perampanel in vivo attenuated brain edema, preserved neurological function, inhibited apoptosis and microglia activation after TBI. Furthermore, perampanel increased the expression of Sirt3 and preserved BBB function after TBI. The effect of perampanel on BBB function and brain edema was abolished by knockdown of Sirt3 in vivo. CONCLUSION: Our results indicate that the noncompetitive AMPAR antagonist perampanel protects the NVU system and reduces brain damage after TBI via activating the Sirt3 cascades.


Subject(s)
Blood-Brain Barrier/metabolism , Neuroprotective Agents/pharmacology , Neurovascular Coupling/physiology , Nitriles/pharmacology , Pyridones/pharmacology , Receptors, AMPA/antagonists & inhibitors , Sirtuins/metabolism , Animals , Blood-Brain Barrier/cytology , Blood-Brain Barrier/drug effects , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Coculture Techniques , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Amino Acid Antagonists/therapeutic use , Female , Male , Neuroprotective Agents/therapeutic use , Neurovascular Coupling/drug effects , Nitriles/therapeutic use , Pregnancy , Pyridones/therapeutic use , Rats , Rats, Sprague-Dawley
10.
Acta Neuropathol Commun ; 9(1): 12, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33413694

ABSTRACT

Vascular dysregulation and cholinergic basal forebrain degeneration are both early pathological events in the development of Alzheimer's disease (AD). Acetylcholine contributes to localised arterial dilatation and increased cerebral blood flow (CBF) during neurovascular coupling via activation of endothelial nitric oxide synthase (eNOS). Decreased vascular reactivity is suggested to contribute to impaired clearance of ß-amyloid (Aß) along intramural periarterial drainage (IPAD) pathways of the brain, leading to the development of cerebral amyloid angiopathy (CAA). However, the possible relationship between loss of cholinergic innervation, impaired vasoreactivity and reduced clearance of Aß from the brain has not been previously investigated. In the present study, intracerebroventricular administration of mu-saporin resulted in significant death of cholinergic neurons and fibres in the medial septum, cortex and hippocampus of C57BL/6 mice. Arterial spin labelling MRI revealed a loss of CBF response to stimulation of eNOS by the Rho-kinase inhibitor fasudil hydrochloride in the cortex of denervated mice. By contrast, the hippocampus remained responsive to drug treatment, in association with altered eNOS expression. Fasudil hydrochloride significantly increased IPAD in the hippocampus of both control and saporin-treated mice, while increased clearance from the cortex was only observed in control animals. Administration of mu-saporin in the TetOAPPSweInd mouse model of AD was associated with a significant and selective increase in Aß40-positive CAA. These findings support the importance of the interrelationship between cholinergic innervation and vascular function in the aetiology and/or progression of CAA and suggest that combined eNOS/cholinergic therapies may improve the efficiency of Aß removal from the brain and reduce its deposition as CAA.


Subject(s)
Acetylcholine/metabolism , Amyloid beta-Peptides/metabolism , Cerebral Amyloid Angiopathy/physiopathology , Cerebral Cortex/blood supply , Cerebrovascular Circulation/physiology , Cholinergic Fibers/physiology , Cholinergic Neurons/physiology , Hippocampus/blood supply , Nitric Oxide Synthase Type III/metabolism , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Animals , Cerebral Amyloid Angiopathy/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebrovascular Circulation/drug effects , Cholinergic Fibers/drug effects , Cholinergic Fibers/metabolism , Cholinergic Neurons/drug effects , Cholinergic Neurons/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Mice , Neurovascular Coupling/drug effects , Neurovascular Coupling/physiology , Saporins/toxicity , Septal Nuclei , Vasodilator Agents/pharmacology
11.
J Cereb Blood Flow Metab ; 41(4): 805-818, 2021 04.
Article in English | MEDLINE | ID: mdl-32538282

ABSTRACT

Local changes in cerebral blood flow are thought to match changes in neuronal activity, a phenomenon termed neurovascular coupling. Hypoxia increases global resting cerebral blood flow, but regional cerebral blood flow (rCBF) changes are non-uniform. Hypoxia decreases baseline rCBF to the default mode network (DMN), which could reflect either decreased neuronal activity or altered neurovascular coupling. To distinguish between these hypotheses, we characterized the effects of hypoxia on baseline rCBF, task performance, and the hemodynamic (BOLD) response to task activity. During hypoxia, baseline CBF increased across most of the brain, but decreased in DMN regions. Performance on memory recall and motion detection tasks was not diminished, suggesting task-relevant neuronal activity was unaffected. Hypoxia reversed both positive and negative task-evoked BOLD responses in the DMN, suggesting hypoxia reverses neurovascular coupling in the DMN of healthy adults. The reversal of the BOLD response was specific to the DMN. Hypoxia produced modest increases in activations in the visual attention network (VAN) during the motion detection task, and had no effect on activations in the visual cortex during visual stimulation. This regional specificity may be particularly pertinent to clinical populations characterized by hypoxemia and may enhance understanding of regional specificity in neurodegenerative disease pathology.


Subject(s)
Default Mode Network/drug effects , Hypoxia, Brain/psychology , Neurovascular Coupling/drug effects , Attention , Cerebrovascular Circulation , Cross-Over Studies , Double-Blind Method , Female , Hemodynamics , Humans , Hypoxia, Brain/diagnostic imaging , Magnetic Resonance Imaging , Male , Mental Recall , Motion Perception , Nerve Net/physiopathology , Neurons , Photic Stimulation , Psychomotor Performance , Visual Cortex/physiopathology , Young Adult
12.
Int J Neurosci ; 131(3): 264-278, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32125198

ABSTRACT

Purpose: Cerebral ischemic stroke, caused by obstruction of the blood flow to the brain, initiates a complex cascade of pathophysiological changes. The aim of the present study was to assess the protective role and the underlying mechanism of troxerutin and cerebroprotein hydrolysate (TCH) injections for five days in rats subjected to middle cerebral artery occlusion (MCAO).Materials and Methods: Male Sprague-Dawley rats treated with either TCH or a vehicle (0.9% saline) via intraperitoneal injection were examined one or three days after MCAO.Results: TCH alleviated neurological deficits and reduced infarct volume, innate immune response, blood-brain barrier destruction, and suppressed cell apoptosis. The therapeutic effects of TCH were achieved by diminished neuronal nitric oxide synthase (nNOS) and inducible nitric oxide synthase (iNOS), and increased endothelial nitric oxide synthase (eNOS). Furthermore, L-NAME showed an inhibitory effect against TCH after MCAO on eNOS expression, NO and peroxynitrite production, neurobehavioral score, and infarct volume.Conclusions: The results indicate that injection of TCH has multifaceted neuroprotective effects against MCAO via regulation of the various NOS isoforms.


Subject(s)
Anticoagulants/administration & dosage , Disease Models, Animal , Hydroxyethylrutoside/analogs & derivatives , Infarction, Middle Cerebral Artery/prevention & control , Neuroprotective Agents/administration & dosage , Neurovascular Coupling/drug effects , Animals , Hydroxyethylrutoside/administration & dosage , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Male , Neurovascular Coupling/physiology , Rats , Rats, Sprague-Dawley
13.
J Alzheimers Dis ; 82(s1): S141-S161, 2021.
Article in English | MEDLINE | ID: mdl-33016916

ABSTRACT

The neurovascular unit (NVU) is responsible for synchronizing the energetic demand, vasodynamic changes, and neurochemical and electrical function of the brain through a closed and interdependent interaction of cell components conforming to brain tissue. In this review, we will focus on cyclin-dependent kinase 5 (CDK5) as a molecular pivot, which plays a crucial role in the healthy function of neurons, astrocytes, and the endothelium and is implicated in the cross-talk of cellular adhesion signaling, ion transmission, and cytoskeletal remodeling, thus allowing the individual and interconnected homeostasis of cerebral parenchyma. Then, we discuss how CDK5 overactivation affects the integrity of the NVU in Alzheimer's disease (AD) and cognitive impairment; we emphasize how CDK5 is involved in the excitotoxicity spreading of glutamate and Ca2+ imbalance under acute and chronic injury. Additionally, we present pharmacological and gene therapy strategies for producing partial depletion of CDK5 activity on neurons, astrocytes, or endothelium to recover neuroplasticity and neurotransmission, suggesting that the NVU should be the targeted tissue unit in protective strategies. Finally, we conclude that CDK5 could be effective due to its intervention on astrocytes by its end feet on the endothelium and neurons, acting as an intermediary cell between systemic and central communication in the brain. This review provides integrated guidance regarding the pathogenesis of and potential repair strategies for AD.


Subject(s)
Astrocytes/metabolism , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Cyclin-Dependent Kinase 5/metabolism , Drug Delivery Systems/methods , Gene Silencing/physiology , Neurovascular Coupling/physiology , Animals , Astrocytes/drug effects , Astrocytes/pathology , Clinical Trials as Topic/methods , Gene Silencing/drug effects , Humans , Neurovascular Coupling/drug effects , Protein Kinase Inhibitors/administration & dosage
14.
Neuroimage ; 225: 117457, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33069862

ABSTRACT

Functional MRI responses are localized to the synaptic sites of evoked inhibitory neurons, but it is unknown whether, or by what mechanisms, these neurons initiate functional hyperemia. Here, the neuronal origins of these hemodynamic responses were investigated by fMRI or local field potential and blood flow measurements during topical application of pharmacological agents when GABAergic granule cells in the rat olfactory bulb were synaptically targeted. First, to examine if postsynaptic activation of these inhibitory neurons was required for neurovascular coupling, we applied an NMDA receptor antagonist during cerebral blood volume-weighted fMRI acquisition and found that responses below the drug application site (up to ~1.5 mm) significantly decreased within ~30 min. Similarly, large decreases in granule cell postsynaptic activities and blood flow responses were observed when AMPA or NMDA receptor antagonists were applied. Second, inhibition of nitric oxide synthase preferentially decreased the initial, fast component of the blood flow response, while inhibitors of astrocyte-specific glutamate transporters and vasoactive intestinal peptide receptors did not decrease blood flow responses. Third, inhibition of GABA release with a presynaptic GABAB receptor agonist caused less reduction of neuronal and blood flow responses compared to the postsynaptic glutamate receptor antagonists. In conclusion, local hyperemia by synaptically-evoked inhibitory neurons was primarily driven by their postsynaptic activities, possibly through NMDA receptor-dependent calcium signaling that was not wholly dependent on nitric oxide.


Subject(s)
Brain/diagnostic imaging , Cerebrovascular Circulation/physiology , GABAergic Neurons/physiology , Neurovascular Coupling/physiology , Amino Acid Transport System X-AG/antagonists & inhibitors , Animals , Brain/physiology , Cerebrovascular Circulation/drug effects , Electric Stimulation , Functional Neuroimaging , GABA-B Receptor Agonists , GABAergic Neurons/drug effects , Laser-Doppler Flowmetry , Magnetic Resonance Imaging , Neural Inhibition , Neurovascular Coupling/drug effects , Nitric Oxide Synthase/antagonists & inhibitors , Olfactory Bulb/cytology , Rats , Receptors, AMPA/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, Vasoactive Intestinal Peptide/antagonists & inhibitors
15.
PLoS One ; 15(9): e0238224, 2020.
Article in English | MEDLINE | ID: mdl-32881886

ABSTRACT

OBJECTIVE: We previously showed that MELAS patients have decreased cerebrovascular reactivity (CVR) (p≤ 0.002) and increased cerebral blood flow (CBF) (p<0.0026); changes correlated with disease severity and % mutant mtDNA (inversely for CVR; directly for CBF). We ran a prospective pilot in 3 MELAS sibs (m.3243A>G tRNALeu(UUR)) with variable % mutant blood mtDNA to assess effects of L-Arginine (L-Arg) (single dose and 6-wk steady-state trial) on regional CBF, arterial CVR and neurovascular coupling. METHODS: Patients were studied with 3T MRI using arterial spin labeling (ASL) to measure CBF and changes in % Blood Oxygen Level Dependent (BOLD) signal to changes in arterial partial pressure of CO2 to measure CVR. Task fMRI consisted of an alternating black and white checkerboard to evaluate visual cortex response in MELAS and controls. RESULTS: Following L-Arg, there was restoration of serum Arg (76-230 µM) in MELAS sibs and a trend towards increasing CVR in frontal and corresponding decrease in occipital cortex; CVR was unchanged globally. There was a 29-37% reduction in baseline CBF in one patient following 6 wks of L-Arg. Pre-treatment fMRI activation in response to visual cortex stimulus was markedly decreased in the same patient compared to controls in primary visual striate cortex V1 and extrastriate regions V2 to V5 with a marked increase toward control values following a single dose and 6 wks of L-Arg. CONCLUSION: Proposed "healing" effect may be due to more efficient utilization of energy substrates with increased cellular energy balances and ensuing reduction in signalling pathways that augment flow in the untreated state. CLASSIFICATION OF EVIDENCE: This prospective pilot study provides Class III evidence that oral L-Arginine (100 mg/kg single dose or 100 mg/kg three times daily po X 6 weeks) normalizes resting blood flow from elevated pre-treatment levels in patients with MELAS syndrome, selectively increases their CVR from reduced pre-treatment levels in regions most impaired at the expense of less abnormal regions, and normalizes reduced BOLD fMRI activation in response to visual cortex stimulus. CLINICAL TRIALS.GOV (NIH): NCT01603446.


Subject(s)
Arginine/therapeutic use , Cerebrovascular Circulation/physiology , MELAS Syndrome/drug therapy , Neurovascular Coupling/physiology , Administration, Oral , Adolescent , Arginine/blood , Arginine/pharmacology , Brain/blood supply , Brain/diagnostic imaging , Brain Mapping , Carbon Dioxide/blood , Cerebrovascular Circulation/drug effects , Female , Humans , Magnetic Resonance Imaging , Male , Neurovascular Coupling/drug effects , Ornithine/blood , Oxygen/blood , Pilot Projects , Prospective Studies , Treatment Outcome , Visual Cortex/drug effects , Young Adult
16.
Hypertension ; 75(6): 1464-1474, 2020 06.
Article in English | MEDLINE | ID: mdl-32362228

ABSTRACT

Antihypertensive medications targeting the renin-angiotensin system have lowered the incidence and progression of Alzheimer disease. Understanding how these medications function could lead to novel therapeutic strategies. AT4Rs (angiotensin IV receptors) have been associated with angiotensin receptor blockers' cognitive, cerebrovascular, and neuroinflammatory rescue in Alzheimer disease models. Yet, whether AT4Rs act alone or with AT2Rs remains unknown. Here, we investigated whether AT2Rs contribute to losartan's benefits and whether chronic AT2R activation could mimic angiotensin receptor blocker benefits in transgenic mice overexpressing familial Alzheimer disease mutations of the human APP (amyloid precursor protein). Losartan-treated mice (10 mg/kg per day, drinking water, 7 months) received intracerebroventricular (1 month) administration of vehicle or AT2R antagonist PD123319 (1.6 nmol/day). PD123319 countered losartan's benefits on spatial learning and memory, neurovascular coupling, and hampered those on oxidative stress and nitric oxide bioavailability. PD123319 did not oppose losartan's benefits on short-term memory and vasodilatory function and had no benefit on neuroinflammation or Aß (amyloid ß) pathology. Mice receiving either vehicle or selective AT2R agonist compound 21 (intracerebroventricular: 1 nmol/day, 1 month or drinking water: 10 mg/kg per day, 7 months), showed no improvement in memory, vasodilatory function, or nitric oxide bioavailability. Compound 21 treatment normalized neurovascular coupling, reduced astrogliosis independent of persisting microgliosis, and exacerbated oxidative stress in APP mice. Compound 21 reduced dense core Aß plaques, but not diffuse plaques or Aß species. Our findings suggest that targeting AT2Rs is not an ideal strategy for restoring Aß-related cognitive and cerebrovascular deficits.


Subject(s)
Alzheimer Disease , Imidazoles/pharmacology , Neurovascular Coupling/drug effects , Plaque, Amyloid , Pyridines/pharmacology , Receptor, Angiotensin, Type 2/metabolism , Receptors, Angiotensin/metabolism , Renin-Angiotensin System/drug effects , Sulfonamides/pharmacology , Thiophenes/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Angiotensin II Type 2 Receptor Blockers/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Cognition/drug effects , Disease Models, Animal , Losartan/pharmacology , Mice , Neuroimmunomodulation , Plaque, Amyloid/immunology , Plaque, Amyloid/pathology , Vasodilation/drug effects
17.
Neuroimage ; 215: 116827, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32289456

ABSTRACT

The neurovascular coupling (NVC) connects neuronal activity to hemodynamic responses in the brain. This connection is the basis for the interpretation of functional magnetic resonance imaging data. Despite the central role of this coupling, we lack detailed knowledge about cell-specific contributions and our knowledge about NVC is mainly based on animal experiments performed during anesthesia. Anesthetics are known to affect neuronal excitability, but how this affects the vessel diameters is not known. Due to the high complexity of NVC data, mathematical modeling is needed for a meaningful analysis. However, neither the relevant neuronal subtypes nor the effects of anesthetics are covered by current models. Here, we present a mathematical model including GABAergic interneurons and pyramidal neurons, as well as the effect of an anesthetic agent. The model is consistent with data from optogenetic experiments from both awake and anesthetized animals, and it correctly predicts data from experiments with different pharmacological modulators. The analysis suggests that no downstream anesthetic effects are necessary if one of the GABAergic interneuron signaling pathways include a Michaelis-Menten expression. This is the first example of a quantitative model that includes both the cell-specific contributions and the effect of an anesthetic agent on the NVC.


Subject(s)
Anesthetics/pharmacology , GABAergic Neurons/physiology , Interneurons/physiology , Models, Theoretical , Neurovascular Coupling/physiology , Pyramidal Cells/physiology , Animals , GABAergic Neurons/drug effects , Interneurons/drug effects , Mice , Mice, Transgenic , Neurovascular Coupling/drug effects , Photic Stimulation/methods , Pyramidal Cells/drug effects
18.
Transl Stroke Res ; 11(4): 720-733, 2020 08.
Article in English | MEDLINE | ID: mdl-31898187

ABSTRACT

The homeostasis of the neurovascular unit (NVU) is disrupted after traumatic brain injury (TBI), and therapeutic strategies targeting the NVU would likely improve neurological outcomes after TBI. Sonic Hedgehog (Shh), which is an endogenous activator of the Hedgehog pathway, promotes brain repair in various injuries. In this study, the controlled cortical impact (CCI) was used to establish a moderate TBI model in adult male Sprague-Dawley rats (250-300 g), and the NVU was reconstructed in vitro from the blood-brain barrier (BBB) and neurons to investigate the effects of exogenous Shh protein on TBI. The modified neurological severity scores (mNSS) and Morris water maze tests were used to evaluate the effect of Shh on neurological function after TBI. The effect of Shh on the NVU in vivo was evaluated by detecting the degrees of cerebral edema and neuronal apoptosis. The integrity and permeability of the BBB, the level of inflammatory factors, and the expression of apoptotic proteins were detected to explore the effect of exogenous Shh on the NVU in vitro. The results showed that exogenous Shh reduced cerebral edema and neuronal apoptosis and promoted neurological recovery after TBI in rats. In vitro experiments showed that Shh-induced activation of the Hedgehog pathway promoted stability of the NVU by reducing damage to the tight junction structure and inhibiting the release of inflammatory factors and neuron apoptosis. Based on these results, the Shh-induced activation of the Hedgehog pathway might be a new promising treatment for TBI.


Subject(s)
Brain Injuries, Traumatic/metabolism , Hedgehog Proteins/administration & dosage , Hedgehog Proteins/metabolism , Neurovascular Coupling , Recovery of Function , Animals , Apoptosis/drug effects , Blood-Brain Barrier/drug effects , Brain Edema/prevention & control , Brain Injuries, Traumatic/prevention & control , Cells, Cultured , Disease Models, Animal , Encephalitis/prevention & control , Male , Neurovascular Coupling/drug effects , Rats, Sprague-Dawley , Signal Transduction/drug effects
19.
Neurobiol Dis ; 134: 104644, 2020 02.
Article in English | MEDLINE | ID: mdl-31669735

ABSTRACT

Angiotensin II type 1 receptor antagonists like losartan have been found to lower the incidence and progression to Alzheimer's disease (AD), as well as rescue cognitive and cerebrovascular deficits in AD mouse models. We previously found that co-administration of an angiotensin IV (AngIV) receptor (AT4R) antagonist prevented losartan's benefits, identifying AT4Rs as a possible target to counter AD pathogenesis. Therein, we investigated whether directly targeting AT4Rs could counter AD pathogenesis in a well-characterized mouse model of AD. Wild-type and human amyloid precursor protein (APP) transgenic (J20 line) mice (4.5 months old) received vehicle or AngIV (~1.3 nmol/day, 1 month) intracerebroventricularly via osmotic minipumps. AngIV restored short-term memory, spatial learning and memory in APP mice. AngIV normalized hippocampal AT4R levels, increased hippocampal subgranular zone cellular proliferation and dendritic arborization, and reduced oxidative stress. AngIV rescued whisker-evoked neurovascular coupling, endothelial- and smooth muscle cell-mediated cerebral vasodilatory responses, and cerebrovascular nitric oxide bioavailability. AngIV did not alter blood pressure, neuroinflammation or amyloid-ß (Aß) pathology. These preclinical findings identify AT4R as a promising target to counter Aß-related cognitive and cerebrovascular deficits in AD.


Subject(s)
Alzheimer Disease/pathology , Angiotensin II/analogs & derivatives , Hippocampus/drug effects , Memory/drug effects , Neurovascular Coupling/drug effects , Amyloid beta-Protein Precursor/genetics , Angiotensin II/pharmacology , Animals , Disease Models, Animal , Humans , Infusions, Intraventricular , Mice , Mice, Inbred C57BL , Mice, Transgenic
20.
Exp Neurol ; 325: 113133, 2020 03.
Article in English | MEDLINE | ID: mdl-31770520

ABSTRACT

Therapeutic hypothermia is a potential protective strategy after stroke. The present study evaluated the neurovascular protective potential of pharmacological hypothermia induced by the neurotensin receptor 1 agonist HPI-201 after severe ischemic stroke. Adult C57BL/6 mice were subjected to filament insertion-induced occlusion of the middle cerebral artery (60 min MCAO). HPI-201 was i.p. injected 120 min after the onset of MCAO to initiate and maintain the body temperature at 32-33°C for 6 hrs. The infarct volume, cell death, integrity of the blood brain barrier (BBB) and neurovascular unit (NVU), inflammation, and functional outcomes were evaluated. The hypothermic treatment significantly suppressed the infarct volume and neuronal cell death, accompanied with reduced caspase-3 activation and BAX expression while Bcl-2 increased in the peri-infarct region. The cellular integrity of the BBB and NVU was significantly improved and brain edema was attenuated in HPI-201-treated mice compared to stroke controls. The hypothermic treatment decreased the expression of inflammatory factors including tumor necrosis factor-α (TNF-α), MMP-9, interleukin-1ß (IL-1ß), the M1 microglia markers IL-12 and inducible nitric oxide synthase (iNOS), while increased the M2 marker arginase-1 (Arg-1). Stroke mice received the hypothermic treatment showed lower neurological severity score (NSS), performed significantly better in functional tests, the mortality rate in the hypothermic group was noticeably lower compared with stroke controls. Taken together, HPI-201 induced pharmacological hypothermia is protective for different neurovascular cells after a severely injured brain, mediated by multiple mechanisms.


Subject(s)
Brain/pathology , Hypothermia, Induced/methods , Infarction, Middle Cerebral Artery/pathology , Neuroprotective Agents/pharmacology , Oligopeptides/pharmacology , Animals , Blood-Brain Barrier/drug effects , Brain/drug effects , Mice, Inbred C57BL , Neurotensin/agonists , Neurovascular Coupling/drug effects , Recovery of Function/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...