Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-38316553

ABSTRACT

Astrocytes play an important role in controlling microvascular diameter and regulating local cerebral blood flow (CBF) in several physiological and pathological scenarios. Neurotransmitters released from active neurons evoke Ca2+ increases in astrocytes, leading to the release of vasoactive metabolites of arachidonic acid (AA) from astrocyte endfeet. Synthesis of prostaglandin E2 (PGE2) and epoxyeicosatrienoic acids (EETs) dilate blood vessels while 20-hydroxyeicosatetraenoic acid (20-HETE) constricts vessels. The release of K+ from astrocyte endfeet also contributes to vasodilation or constriction in a concentration-dependent manner. Whether astrocytes exert a vasodilation or vasoconstriction depends on the local microenvironment, including the metabolic status, the concentration of Ca2+ reached in the endfoot, and the resting vascular tone. Astrocytes also contribute to the generation of steady-state vascular tone. Tonic release of both 20-HETE and ATP from astrocytes constricts vascular smooth muscle cells, generating vessel tone, whereas tone-dependent elevations in endfoot Ca2+ produce tonic prostaglandin dilators to limit the degree of constriction. Under pathological conditions, including Alzheimer's disease, epilepsy, stroke, and diabetes, disruption of normal astrocyte physiology can compromise the regulation of blood flow, with negative consequences for neurological function.


Subject(s)
Astrocytes , Cerebrovascular Circulation , Astrocytes/metabolism , Cerebrovascular Circulation/physiology , Neurons , Prostaglandins/metabolism
2.
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
3.
J Neurosci ; 44(2)2024 Jan 10.
Article in English | MEDLINE | ID: mdl-37989594

ABSTRACT

Glutamate spillover from the synapse is tightly regulated by astrocytes, limiting the activation of extrasynaptically located NMDA receptors (NMDAR). The processes of astrocytes are dynamic and can modulate synaptic physiology. Though norepinephrine (NE) and ß-adrenergic receptor (ß-AR) activity can modify astrocyte volume, this has yet to be confirmed outside of sensory cortical areas, nor has the effect of noradrenergic signaling on glutamate spillover and neuronal NMDAR activity been explored. We monitored changes to astrocyte process volume in response to noradrenergic agonists in the medial prefrontal cortex of male and female mice. Both NE and the ß-AR agonist isoproterenol (ISO) increased process volume by ∼20%, significantly higher than changes seen when astrocytes had G-protein signaling blocked by GDPßS. We measured the effect of ß-AR signaling on evoked NMDAR currents. While ISO did not affect single stimulus excitatory currents of Layer 5 pyramidal neurons, ISO reduced NMDAR currents evoked by 10 stimuli at 50 Hz, which elicits glutamate spillover, by 18%. After isolating extrasynaptic NMDARs by blocking synaptic NMDARs with the activity-dependent NMDAR blocker MK-801, ISO similarly reduced extrasynaptic NMDAR currents in response to 10 stimuli by 18%. Finally, blocking ß-AR signaling in the astrocyte network by loading them with GDPßS reversed the ISO effect on 10 stimuli-evoked NMDAR currents. These results demonstrate that astrocyte ß-AR activity reduces extrasynaptic NMDAR recruitment, suggesting that glutamate spillover is reduced.


Subject(s)
Astrocytes , Receptors, N-Methyl-D-Aspartate , Mice , Animals , Male , Female , Receptors, N-Methyl-D-Aspartate/metabolism , Astrocytes/metabolism , Pyramidal Cells/physiology , Prefrontal Cortex/physiology , Glutamic Acid/physiology , Receptors, Adrenergic, beta , Synapses/physiology
4.
Glia ; 70(10): 1927-1937, 2022 10.
Article in English | MEDLINE | ID: mdl-35678626

ABSTRACT

Volume transmission plays an essential role in CNS function, with neurotransmitters released from synapses diffusing through the extracellular space (ECS) to distant sites. Changes in the ECS volume fraction (α) will influence the diffusion and the concentration of transmitters within the ECS. We have recently shown that neuronal activity evoked by physiological photic stimuli results in rapid decreases in ECS α as large as 10% in the retina. We now characterize the cellular mechanisms responsible for this ECS shrinkage. We find that block of inwardly rectifying K+ channels with Ba2+ , inhibition of the Na+ /K+ /2Cl- cotransporter with bumetanide, or block of AQP4 water channels with TGN-020 do not diminish the light-evoked ECS decrease. Inhibition of the Na+ /HCO3 - cotransporter by removing HCO3 - from the superfusate, in contrast, reduces the light-evoked ECS decrease by 95.6%. Inhibition of the monocarboxylate transporter with alpha-cyano-4-hydroxycinnamate (4-CIN) also reduces the ECS shrinkage, but only by 32.5%. We tested whether the swelling of Müller cells, the principal glial cells of the retina, is responsible for the light-evoked ECS shrinkage. Light stimulation evoked a 6.3% increase in the volume of the fine processes of Müller cells. This volume increase was reduced by 97.1% when HCO3 - was removed from the superfusate. We conclude that a large fraction of the activity-dependent decrease in ECS α is generated by the activation of the Na+ /HCO3 - cotransporter in Müller cells. The monocarboxylate transporter may also contribute to the response.


Subject(s)
Extracellular Space , Neuroglia , Bumetanide/pharmacology , Neuroglia/physiology , Neurons , Potassium , Retina , Sodium
5.
J Cereb Blood Flow Metab ; 42(8): 1534-1546, 2022 08.
Article in English | MEDLINE | ID: mdl-35296178

ABSTRACT

Hypoglycemia triggers increases in cerebral blood flow (CBF), augmenting glucose supply to the brain. We have tested whether astrocytes, which can regulate vessel tone, contribute to this CBF increase. We hypothesized that hypoglycemia-induced adenosine signaling acts to increase astrocyte Ca2+ activity, which then causes the release of prostaglandins (PGs) and epoxyeicosatrienoic acids (EETs), leading to the dilation of brain arterioles and blood flow increases. We used an awake mouse model to investigate the effects of insulin-induced hypoglycemia on arterioles and astrocytes in the somatosensory cortex. During insulin-induced hypoglycemia, penetrating arterioles dilated and astrocyte Ca2+ signaling increased when blood glucose dropped below a threshold of ∼50 mg/dL. Application of the A2A adenosine receptor antagonist ZM-241385 eliminated hypoglycemia-evoked astrocyte Ca2+ increases and reduced arteriole dilations by 44% (p < 0.05). SC-560 and miconazole, which block the production of the astrocyte vasodilators PGs and EETs respectively, reduced arteriole dilations in response to hypoglycemia by 89% (p < 0.001) and 76% (p < 0.001). Hypoglycemia-induced arteriole dilations were decreased by 65% (p < 0.001) in IP3R2 knockout mice, which have reduced astrocyte Ca2+ signaling compared to wild-type. These results support the hypothesis that astrocytes contribute to hypoglycemia-induced increases in CBF by releasing vasodilators in a Ca2+-dependent manner.


Subject(s)
Hypoglycemia , Insulins , Animals , Arterioles/metabolism , Astrocytes/metabolism , Cerebrovascular Circulation/physiology , Hypoglycemia/metabolism , Insulins/metabolism , Insulins/pharmacology , Mice , Vasodilator Agents/pharmacology
6.
Glia ; 70(3): 508-521, 2022 03.
Article in English | MEDLINE | ID: mdl-34767261

ABSTRACT

The brain requires an adequate supply of oxygen and nutrients to maintain proper function as neuronal activity varies. This is achieved, in part, through neurovascular coupling mechanisms that mediate local increases in blood flow through the dilation of arterioles and capillaries. The role of astrocytes in mediating this functional hyperemia response is controversial. Specifically, the function of astrocyte Ca2+ signaling is unclear. Cortical arterioles dilate in the absence of astrocyte Ca2+ signaling, but previous work suggests that Ca2+ increases are necessary for capillary dilation. This question has not been fully addressed in vivo, however, and we have reexamined the role of astrocyte Ca2+ signaling in vessel dilation in the barrel cortex of awake, behaving mice. We recorded evoked vessel dilations and astrocyte Ca2+ signaling in response to whisker stimulation. Experiments were carried out on WT and IP3R2 KO mice, a transgenic model where astrocyte Ca2+ signaling is substantially reduced. Compared to WT mice at rest, Ca2+ signaling in astrocyte endfeet contacting capillaries increased by 240% when whisker stimulation evoked running. In contrast, Ca2+ signaling was reduced to 9% of WT values in IP3R2 KO mice. In all three conditions, however, the amplitude of capillary dilation was largely unchanged. In addition, the latency to the onset of astrocyte Ca2+ signaling lagged behind dilation onset in most trials, although a subset of rapid onset Ca2+ events with latencies as short as 0.15 s occurred. In summary, we found that whisker stimulation-evoked capillary dilations occurred independent of astrocyte Ca2+ increases in the cerebral cortex.


Subject(s)
Astrocytes , Calcium Signaling , Animals , Astrocytes/metabolism , Calcium/metabolism , Calcium Signaling/physiology , Capillaries/metabolism , Cerebral Cortex/metabolism , Dilatation , Mice
7.
J Neurosci ; 40(41): 7785-7794, 2020 10 07.
Article in English | MEDLINE | ID: mdl-32887746

ABSTRACT

The extracellular space (ECS) plays an important role in the physiology of neural circuits. Despite our detailed understanding of the cellular architecture of the mammalian retina, little is known about the organization and dynamics of the retinal ECS. We developed an optical technique based on two-photon imaging of fluorescently labeled extracellular fluid to measure the ECS volume fraction (α) in the ex vivo retina of male and female mice. This method has high spatial resolution and can detect rapid changes in α evoked by osmotic challenge and neuronal activity. The measured ECS α varied dramatically in different layers of the adult mouse retina, with α equaling ∼0.050 in the ganglion cell layer, ∼0.122 in the inner plexiform layer (IPL), ∼0.025 in the inner nuclear layer (INL), ∼0.087 in the outer plexiform layer, and ∼0.026 in the outer nuclear layer (ONL). ECS α was significantly larger early in retinal development; α was 67% larger in the IPL and 100% larger in the INL in neonatal mice compared with adults. In adult retinas, light stimulation evoked rapid decreases in ECS α. Light-driven reductions in ECS α were largest in the IPL, where visual stimuli decreased α values ∼10%. These light-evoked decreases demonstrate that a physiological stimulus can lead to rapid changes in ECS α and indicate that activity-dependent regulation of extracellular space may contribute to visual processing in the retina.SIGNIFICANCE STATEMENT The volume fraction of the extracellular space (ECS α), that portion of CNS tissue occupied by interstitial space, influences the diffusion of neurotransmitters from the synaptic cleft and the volume transmission of transmitters. However, ECS α has never been measured in live retina, and little is known about how ECS α varies following physiological stimulation. Here we show that ECS α values vary dramatically between different retinal layers and decrease by 10% following light stimulation. ECS α differences within the retina will influence volume transmission and light-evoked α variations may modulate synaptic transmission and visual processing in the retina. Activity-dependent ECS α variations may represent a mechanism of synaptic modulation throughout the CNS.


Subject(s)
Extracellular Space/physiology , Retina/ultrastructure , Absorptiometry, Photon , Animals , Animals, Newborn , Extracellular Space/radiation effects , Female , Fluorescent Dyes , Male , Mice , Mice, Inbred C57BL , Neural Pathways/physiology , Neural Pathways/ultrastructure , Neurons/physiology , Neurons/ultrastructure , Osmotic Pressure , Photic Stimulation , Retina/growth & development , Retina/physiology , Retinal Ganglion Cells/physiology , Retinal Ganglion Cells/ultrastructure
8.
Vis Neurosci ; 37: E004, 2020 07 20.
Article in English | MEDLINE | ID: mdl-32684188

ABSTRACT

Blood flow in the retina increases in response to light-evoked neuronal activity, ensuring that retinal neurons receive an adequate supply of oxygen and nutrients as metabolic demands vary. This response, termed "functional hyperemia," is disrupted in diabetic retinopathy. The reduction in functional hyperemia may result in retinal hypoxia and contribute to the development of retinopathy. This review will discuss the neurovascular coupling signaling mechanisms that generate the functional hyperemia response in the retina, the changes to neurovascular coupling that occur in diabetic retinopathy, possible treatments for restoring functional hyperemia and retinal oxygen levels, and changes to functional hyperemia that occur in the diabetic brain.


Subject(s)
Diabetic Retinopathy/physiopathology , Regional Blood Flow/physiology , Retinal Vessels/physiopathology , Animals , Blood Flow Velocity , Humans , Hyperemia/physiopathology , Oxygen/blood
9.
J Neurosurg ; 132(6): 1820-1828, 2019 Apr 05.
Article in English | MEDLINE | ID: mdl-30952117

ABSTRACT

OBJECTIVE: Cortical spreading depolarization (CSD) has been linked to poor clinical outcomes in the setting of traumatic brain injury, malignant stroke, and subarachnoid hemorrhage. There is evidence that electrocautery during neurosurgical procedures can also evoke CSD waves in the brain. It is unknown whether blood contacting the cortical surface during surgical bleeding affects the frequency of spontaneous or surgery-induced CSDs. Using a mouse neurosurgical model, the authors tested the hypothesis that electrocautery can induce CSD waves and that surgical field blood (SFB) is associated with more CSDs. The authors also investigated whether CSD can be reliably observed by monitoring the fluorescence of GCaMP6f expressed in neurons. METHODS: CSD waves were monitored by using confocal microscopy to detect fluorescence increases at the cortical surface in mice expressing GCaMP6f in CamKII-positive neurons. The cortical surface was electrocauterized through an adjacent burr hole. SFB was simulated by applying a drop of tail vein blood to the brain through the same burr hole. RESULTS: CSD waves were readily detected in GCaMP6f-expressing mice. Monitoring GCaMP6f fluorescence provided far better sensitivity and spatial resolution than detecting CSD events by observing changes in the intrinsic optical signal (IOS). Forty-nine percent of the CSD waves identified by GCaMP6f had no corresponding IOS signal. Electrocautery evoked CSD waves. On average, 0.67 ± 0.08 CSD events were generated per electrocautery episode, and multiple CSD waves could be induced in the same mouse by repeated cauterization (average, 7.9 ± 1.3 events; maximum number in 1 animal, 13 events). In the presence of SFB, significantly more spontaneous CSDs were generated (1.35 ± 0.37 vs 0.13 ± 0.16 events per hour, p = 0.002). Ketamine effectively decreased the frequency of spontaneous CSD waves (1.35 ± 0.37 to 0.36 ± 0.15 CSD waves per hour, p = 0.016) and electrocautery-stimulated CSD waves (0.80 ± 0.05 to 0.18 ± 0.08 CSD waves per electrocautery, p = 0.00002). CONCLUSIONS: CSD waves are detected with far greater sensitivity and fidelity by monitoring GCaMP6f signals in neurons than by monitoring IOSs. Electrocautery reliably evokes CSD waves, and the frequency of spontaneous CSD waves is increased when blood is applied to the cortical surface. These experimental conditions recapitulate common scenarios in the neurosurgical operating room. Ketamine, a clinically available pharmaceutical agent, can block stimulated and spontaneous CSDs. More research is required to understand the clinical importance of intraoperative CSD.

10.
Neuron ; 99(2): 248-250, 2018 07 25.
Article in English | MEDLINE | ID: mdl-30048612

ABSTRACT

Cerebral blood flow increases in regions of increased brain activity. In this issue of Neuron, Rungta et al. (2018) characterize the contribution of different vascular compartments in generating this increase and outline the time course of arteriole and capillary dilation in generating functional hyperemia.


Subject(s)
Capillaries , Hyperemia , Arterioles , Brain , Humans , Synapses
11.
Neuroscientist ; 24(1): 73-83, 2018 02.
Article in English | MEDLINE | ID: mdl-28403673

ABSTRACT

Neuronal activity within the brain evokes local increases in blood flow, a response termed functional hyperemia. This response ensures that active neurons receive sufficient oxygen and nutrients to maintain tissue function and health. In this review, we discuss the functions of functional hyperemia, the types of vessels that generate the response, and the signaling mechanisms that mediate neurovascular coupling, the communication between neurons and blood vessels. Neurovascular coupling signaling is mediated primarily by the vasoactive metabolites of arachidonic acid (AA), by nitric oxide, and by K+. While much is known about these pathways, many contentious issues remain. We highlight two controversies, the role of glial cell Ca2+ signaling in mediating neurovascular coupling and the importance of capillaries in generating functional hyperemia. We propose signaling pathways that resolve these controversies. In this scheme, capillary dilations are generated by Ca2+ increases in astrocyte endfeet, leading to production of AA metabolites. In contrast, arteriole dilations are generated by Ca2+ increases in neurons, resulting in production of nitric oxide and AA metabolites. Arachidonic acid from neurons also diffuses into astrocyte endfeet where it is converted into additional vasoactive metabolites. While this scheme resolves several discrepancies in the field, many unresolved challenges remain and are discussed in the final section of the review.


Subject(s)
Brain/physiopathology , Cerebrovascular Disorders/physiopathology , Hyperemia/physiopathology , Animals , Humans
12.
J Neurosci ; 36(36): 9435-45, 2016 09 07.
Article in English | MEDLINE | ID: mdl-27605617

ABSTRACT

UNLABELLED: The brain is critically dependent on the regulation of blood flow to nourish active neurons. One widely held hypothesis of blood flow regulation holds that active neurons stimulate Ca(2+) increases in glial cells, triggering glial release of vasodilating agents. This hypothesis has been challenged, as arteriole dilation can occur in the absence of glial Ca(2+) signaling. We address this controversy by imaging glial Ca(2+) signaling and vessel dilation in the mouse retina. We find that sensory stimulation results in Ca(2+) increases in the glial endfeet contacting capillaries, but not arterioles, and that capillary dilations often follow spontaneous Ca(2+) signaling. In IP3R2(-/-) mice, where glial Ca(2+) signaling is reduced, light-evoked capillary, but not arteriole, dilation is abolished. The results show that, independent of arterioles, capillaries actively dilate and regulate blood flow. Furthermore, the results demonstrate that glial Ca(2+) signaling regulates capillary but not arteriole blood flow. SIGNIFICANCE STATEMENT: We show that a Ca(2+)-dependent glial cell signaling mechanism is responsible for regulating capillary but not arteriole diameter. This finding resolves a long-standing controversy regarding the role of glial cells in regulating blood flow, demonstrating that glial Ca(2+) signaling is both necessary and sufficient to dilate capillaries. While the relative contributions of capillaries and arterioles to blood flow regulation remain unclear, elucidating the mechanisms that regulate capillary blood flow may ultimately lead to the development of therapies for treating diseases where blood flow regulation is disrupted, including Alzheimer's disease, stroke, and diabetic retinopathy. This finding may also aid in revealing the underlying neuronal activity that generates BOLD fMRI signals.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Capillaries/physiology , Ependymoglial Cells/physiology , Regional Blood Flow/physiology , Retina/cytology , Animals , Antigens/metabolism , Calcium Signaling/genetics , Capillaries/drug effects , Ependymoglial Cells/drug effects , Excitatory Amino Acid Transporter 1/genetics , Excitatory Amino Acid Transporter 1/metabolism , Female , Glial Fibrillary Acidic Protein/metabolism , Glycosaminoglycans/physiology , In Vitro Techniques , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Proteoglycans/metabolism , Regional Blood Flow/drug effects , Transcription Factors/genetics , Transcription Factors/metabolism , Visual Pathways/physiology
13.
J Cereb Blood Flow Metab ; 36(9): 1579-91, 2016 09.
Article in English | MEDLINE | ID: mdl-27389181

ABSTRACT

Cortical spreading depolarization is a metabolically costly phenomenon that affects the brain in both health and disease. Following severe stroke, subarachnoid hemorrhage, or traumatic brain injury, cortical spreading depolarization exacerbates tissue damage and enlarges infarct volumes. It is not known, however, whether spreading depolarization also occurs in the retina in vivo. We report now that spreading depolarization episodes are generated in the in vivo rat retina following retinal vessel occlusion produced by photothrombosis. The properties of retinal spreading depolarization are similar to those of cortical spreading depolarization. Retinal spreading depolarization waves propagate at a velocity of 3.0 ± 0.1 mm/min and are associated with a negative shift in direct current potential, a transient cessation of neuronal spiking, arteriole constriction, and a decrease in tissue O2 tension. The frequency of retinal spreading depolarization generation in vivo is reduced by administration of the NMDA antagonist MK-801 and the 5-HT(1D) agonist sumatriptan. Branch retinal vein occlusion is a leading cause of vision loss from vascular disease. Our results suggest that retinal spreading depolarization could contribute to retinal damage in acute retinal ischemia and demonstrate that pharmacological agents can reduce retinal spreading depolarization frequency after retinal vessel occlusion. Blocking retinal spreading depolarization generation may represent a therapeutic strategy for preserving vision in branch retinal vein occlusion patients.


Subject(s)
Brain Ischemia/physiopathology , Cortical Spreading Depression , Retina/physiopathology , Animals , Dizocilpine Maleate/administration & dosage , Dizocilpine Maleate/therapeutic use , Excitatory Amino Acid Antagonists , Rats , Retina/injuries , Retinal Artery Occlusion/drug therapy , Serotonin 5-HT1 Receptor Agonists , Sumatriptan/administration & dosage , Sumatriptan/therapeutic use
14.
Sci Rep ; 6: 21075, 2016 Feb 08.
Article in English | MEDLINE | ID: mdl-26852722

ABSTRACT

The aetiology of diabetic retinopathy (DR), the leading cause of blindness in the developed world, remains controversial. One hypothesis holds that retinal hypoxia, exacerbated by the high O2 consumption of rod photoreceptors in the dark, is a primary cause of DR. Based on this prediction we investigated whether early retinal abnormalities in streptozotocin-induced diabetic rats are alleviated by preventing the rods from dark adapting. Diabetic rats and their non-diabetic littermates were housed in a 12:12 hour light-dim light photocycle (30 lux during the day and 3 lux at night). Progression of early retinal abnormalities in diabetic rats was assessed by monitoring the ERG b-wave and oscillatory potentials, Müller cell reactive gliosis, and neuronal cell death, as assayed by TUNEL staining and retinal thickness at 6 and 12 weeks after diabetes induction. Maintaining diabetic animals in a dim-adapting light did not slow the progression of these neuronal and glial changes when compared to diabetic rats maintained in a standard 12:12 hour light-dark photocycle (30 lux during the day and 0 lux at night). Our results indicate that neuronal and glial abnormalities in early stages of diabetes are not exacerbated by rod photoreceptor O2 consumption in the dark.


Subject(s)
Cell Hypoxia/physiology , Dark Adaptation , Diabetes Mellitus, Experimental/pathology , Diabetic Retinopathy/etiology , Oxygen Consumption/physiology , Retina/pathology , Adaptation, Ocular , Animals , Diabetes Mellitus, Experimental/metabolism , Disease Progression , Electroretinography , Light , Male , Neuroglia/pathology , Rats , Rats, Sprague-Dawley , Retina/metabolism , Streptozocin
15.
eNeuro ; 2(2)2015.
Article in English | MEDLINE | ID: mdl-26082942

ABSTRACT

Blood flow is a useful indicator of the metabolic state of the retina. However, accurate measurement of retinal blood flow is difficult to achieve in practice. Most existing optical techniques used for measuring blood flow require complex assumptions and calculations. We describe here a simple and direct method for calculating absolute blood flow in vessels of all sizes in the rat retina. The method relies on ultrafast confocal line scans to track the passage of fluorescently labeled red blood cells (fRBCs). The accuracy of the blood flow measurements was verified by (1) comparing blood flow calculated independently using either flux or velocity combined with diameter measurements, (2) measuring total retinal blood flow in arterioles and venules, (3) measuring blood flow at vessel branch points, and (4) measuring changes in blood flow in response to hyperoxic and hypercapnic challenge. Confocal line scans oriented parallel and diagonal to vessels were used to compute fRBC velocity and to examine velocity profiles across the width of vessels. We demonstrate that these methods provide accurate measures of absolute blood flow and velocity in retinal vessels of all sizes.

16.
Philos Trans R Soc Lond B Biol Sci ; 370(1672)2015 Jul 05.
Article in English | MEDLINE | ID: mdl-26009774

ABSTRACT

Astrocytes in the brain release transmitters that actively modulate neuronal excitability and synaptic efficacy. Astrocytes also release vasoactive agents that contribute to neurovascular coupling. As reviewed in this article, Müller cells, the principal retinal glial cells, modulate neuronal activity and blood flow in the retina. Stimulated Müller cells release ATP which, following its conversion to adenosine by ectoenzymes, hyperpolarizes retinal ganglion cells by activation of A1 adenosine receptors. This results in the opening of G protein-coupled inwardly rectifying potassium (GIRK) channels and small conductance Ca(2+)-activated K(+) (SK) channels. Tonic release of ATP also contributes to the generation of tone in the retinal vasculature by activation of P2X receptors on vascular smooth muscle cells. Vascular tone is lost when glial cells are poisoned with the gliotoxin fluorocitrate. The glial release of vasoactive metabolites of arachidonic acid, including prostaglandin E2 (PGE2) and epoxyeicosatrienoic acids (EETs), contributes to neurovascular coupling in the retina. Neurovascular coupling is reduced when neuronal stimulation of glial cells is interrupted and when the synthesis of arachidonic acid metabolites is blocked. Neurovascular coupling is compromised in diabetic retinopathy owing to the loss of glial-mediated vasodilation. This loss can be reversed by inhibiting inducible nitric oxide synthase. It is likely that future research will reveal additional important functions of the release of transmitters from glial cells.


Subject(s)
Ependymoglial Cells/physiology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Neurotransmitter Agents/metabolism , Regional Blood Flow/physiology , Retinal Neurons/physiology , Retinal Vessels/physiology , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Action Potentials/physiology , Ependymoglial Cells/metabolism , Humans
17.
Article in English | MEDLINE | ID: mdl-25818565

ABSTRACT

Neuronal activity results in increased blood flow in the brain, a response named functional hyperemia. Astrocytes play an important role in mediating this response. Neurotransmitters released from active neurons evoke Ca(2+) increases in astrocytes, leading to the release of vasoactive metabolites of arachidonic acid from astrocyte endfeet onto blood vessels. Synthesis of prostaglandin E2 (PGE2) and epoxyeicosatrienoic acids (EETs) dilate blood vessels, whereas 20-hydroxyeicosatetraenoic acid (20-HETE) constricts vessels. The release of K(+) from astrocyte endfeet may also contribute to vasodilation. Oxygen modulates astrocyte regulation of blood flow. Under normoxic conditions, astrocytic Ca(2+) signaling results in vasodilation, whereas under hyperoxic conditions, vasoconstriction is favored. Astrocytes also contribute to the generation of vascular tone. Tonic release of both 20-HETE and ATP from astrocytes constricts vascular smooth muscle cells, generating vessel tone. Under pathological conditions, including Alzheimer's disease and diabetic retinopathy, disruption of normal astrocyte physiology can compromise the regulation of blood flow.


Subject(s)
Astrocytes/physiology , Cerebrovascular Circulation/physiology , Brain/blood supply , Brain/cytology , Brain/physiology , Capillaries/physiology , Humans
18.
J Neurosci ; 34(34): 11504-13, 2014 Aug 20.
Article in English | MEDLINE | ID: mdl-25143628

ABSTRACT

Light stimulation evokes neuronal activity in the retina, resulting in the dilation of retinal blood vessels and increased blood flow. This response, named functional hyperemia, brings oxygen and nutrients to active neurons. However, it remains unclear which vessels mediate functional hyperemia. We have characterized blood flow regulation in the rat retina in vivo by measuring changes in retinal vessel diameter and red blood cell (RBC) flux evoked by a flickering light stimulus. We found that, in first- and second-order arterioles, flicker evoked large (7.5 and 5.0%), rapid (0.73 and 0.70 s), and consistent dilations. Flicker-evoked dilations in capillaries were smaller (2.0%) and tended to have a slower onset (0.97 s), whereas dilations in venules were smaller (1.0%) and slower (1.06 s) still. The proximity of pericyte somata did not predict capillary dilation amplitude. Expression of the contractile protein α-smooth muscle actin was high in arterioles and low in capillaries. Unexpectedly, we found that blood flow in the three vascular layers was differentially regulated. Flicker stimulation evoked far larger dilations and RBC flux increases in the intermediate layer capillaries than in the superficial and deep layer capillaries (2.6 vs 0.9 and 0.7% dilation; 25.7 vs 0.8 and 11.3% RBC flux increase). These results indicate that functional hyperemia in the retina is driven primarily by active dilation of arterioles. The dilation of intermediate layer capillaries is likely mediated by active mechanisms as well. The physiological consequences of differential regulation in the three vascular layers are discussed.


Subject(s)
Microvessels/physiology , Regional Blood Flow/physiology , Retina/anatomy & histology , Retinal Vessels/physiology , Acetamides/metabolism , Actins/metabolism , Analysis of Variance , Animals , Antigens/metabolism , Dextrans/metabolism , Erythrocytes/physiology , Flicker Fusion , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/metabolism , Lectins/metabolism , Male , Microscopy, Confocal , Proteoglycans/metabolism , Rats , Rats, Sprague-Dawley , Retinal Vessels/cytology , Time Factors
19.
J Physiol ; 592(3): 491-504, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24277867

ABSTRACT

Purinergic control of vascular tone in the CNS has been largely unexplored. This study examines the contribution of endogenous extracellular ATP, acting on vascular smooth muscle cells, in controlling vascular tone in the in vivo rat retina. Retinal vessels were labelled by i.v. injection of a fluorescent dye and imaged with scanning laser confocal microscopy. The diameters of primary arterioles were monitored under control conditions and following intravitreal injection of pharmacological agents. Apyrase (500 units ml(-1)), an ATP hydrolysing enzyme, dilated retinal arterioles by 40.4 ± 2.8%, while AOPCP (12.5 mm), an ecto-5'-nucleotidase inhibitor that increases extracellular ATP levels, constricted arterioles by 58.0 ± 3.8% (P < 0.001 for both), demonstrating the importance of ATP in the control of basal vascular tone. Suramin (500 µm), a broad-spectrum P2 receptor antagonist, dilated retinal arterioles by 50.9 ± 3.7% (P < 0.001). IsoPPADS (300 µm) and TNP-ATP (50 µm), more selective P2X antagonists, dilated arterioles by 41.0 ± 5.3% and 55.2 ± 6.1% respectively (P < 0.001 for both). NF023 (50 µm), a potent antagonist of P2X1 receptors, dilated retinal arterioles by 32.1 ± 2.6% (P < 0.001). A438079 (500 µm) and AZ10606120 (50 µm), P2X7 antagonists, had no effect on basal vascular tone (P = 0.99 and P = 1.00 respectively). In the ex vivo retina, the P2X1 receptor agonist α,ß-methylene ATP (300 nm) evoked sustained vasoconstrictions of 18.7 ± 3.2% (P < 0.05). In vivo vitreal injection of the gliotoxin fluorocitrate (150 µm) dilated retinal vessels by 52.3 ± 1.1% (P < 0.001) and inhibited the vasodilatory response to NF023 (50 µm, 7.9 ± 2.0%; P < 0.01). These findings suggest that vascular tone in rat retinal arterioles is maintained by tonic release of ATP from the retina. ATP acts on P2X1 receptors, although contributions from other P2X and P2Y receptors cannot be ruled out. Retinal glial cells are a possible source of the vasoconstricting ATP.


Subject(s)
Purinergic P2X Receptor Agonists/pharmacology , Purinergic P2X Receptor Antagonists/pharmacology , Retinal Artery/metabolism , Vasodilation , Adenosine Triphosphate/metabolism , Animals , Apyrase/pharmacology , Arterioles/drug effects , Arterioles/metabolism , Arterioles/physiology , Male , Rats , Rats, Long-Evans , Retinal Artery/drug effects , Retinal Artery/physiology
20.
J Undergrad Neurosci Educ ; 12(1): A11-7, 2013.
Article in English | MEDLINE | ID: mdl-24319385

ABSTRACT

MetaNeuron, a neuron simulation program, is an effective interactive tool for teaching cellular neurophysiology. The computer program simulates a wide range of neuronal behavior in its six lessons: i) Resting Membrane Potential, ii) Membrane Time Constant, iii) Membrane Length Constant, iv) Axon Action Potential, v) Axon Voltage Clamp, and vi) Synaptic Potential. The program is designed foremost as a platform for conducting neurophysiology experiments in silico. Neuronal parameters are easily modified and a virtual stimulator injects single or double current pulses into the neuron. Phenomena such as temporal summation of synaptic potentials, passive spread of a synaptic potential from the dendrite to the soma, the refractory period, families of voltage-clamp traces, and the reversal potential of synaptic responses, are easily illustrated in MetaNeuron. Responses are displayed graphically and can be measured with a cursor. Families of traces can be generated and viewed in rotatable 3D plots. Mac and Windows versions of the program can be downloaded, free of charge, onto individual student computers from the website www.MetaNeuron.org. A manual containing operating instructions, a description of the lessons, and exercises conducted on MetaNeuron, can also be downloaded for free.

SELECTION OF CITATIONS
SEARCH DETAIL
...