Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
J Neurosci ; 44(2)2024 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-37989594

RESUMEN

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.


Asunto(s)
Astrocitos , Receptores de N-Metil-D-Aspartato , Ratones , Animales , Masculino , Femenino , Receptores de N-Metil-D-Aspartato/metabolismo , Astrocitos/metabolismo , Células Piramidales/fisiología , Corteza Prefrontal/fisiología , Ácido Glutámico/fisiología , Receptores Adrenérgicos beta , Sinapsis/fisiología
2.
Glia ; 70(10): 1927-1937, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35678626

RESUMEN

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.


Asunto(s)
Espacio Extracelular , Neuroglía , Bumetanida/farmacología , Neuroglía/fisiología , Neuronas , Potasio , Retina , Sodio
3.
Glia ; 70(3): 508-521, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34767261

RESUMEN

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.


Asunto(s)
Astrocitos , Señalización del Calcio , Animales , Astrocitos/metabolismo , Calcio/metabolismo , Señalización del Calcio/fisiología , Capilares/metabolismo , Corteza Cerebral/metabolismo , Dilatación , Ratones
4.
J Neurosci ; 40(41): 7785-7794, 2020 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-32887746

RESUMEN

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.


Asunto(s)
Espacio Extracelular/fisiología , Retina/ultraestructura , Absorciometría de Fotón , Animales , Animales Recién Nacidos , Espacio Extracelular/efectos de la radiación , Femenino , Colorantes Fluorescentes , Masculino , Ratones , Ratones Endogámicos C57BL , Vías Nerviosas/fisiología , Vías Nerviosas/ultraestructura , Neuronas/fisiología , Neuronas/ultraestructura , Presión Osmótica , Estimulación Luminosa , Retina/crecimiento & desarrollo , Retina/fisiología , Células Ganglionares de la Retina/fisiología , Células Ganglionares de la Retina/ultraestructura
5.
Vis Neurosci ; 37: E004, 2020 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-32684188

RESUMEN

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.


Asunto(s)
Retinopatía Diabética/fisiopatología , Flujo Sanguíneo Regional/fisiología , Vasos Retinianos/fisiopatología , Animales , Velocidad del Flujo Sanguíneo , Humanos , Hiperemia/fisiopatología , Oxígeno/sangre
6.
J Neurosci ; 36(36): 9435-45, 2016 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-27605617

RESUMEN

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.


Asunto(s)
Señalización del Calcio/fisiología , Calcio/metabolismo , Capilares/fisiología , Células Ependimogliales/fisiología , Flujo Sanguíneo Regional/fisiología , Retina/citología , Animales , Antígenos/metabolismo , Señalización del Calcio/genética , Capilares/efectos de los fármacos , Células Ependimogliales/efectos de los fármacos , Transportador 1 de Aminoácidos Excitadores/genética , Transportador 1 de Aminoácidos Excitadores/metabolismo , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Glicosaminoglicanos/fisiología , Técnicas In Vitro , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteoglicanos/metabolismo , Flujo Sanguíneo Regional/efectos de los fármacos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Vías Visuales/fisiología
7.
Nature ; 468(7321): 232-43, 2010 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-21068832

RESUMEN

Blood flow in the brain is regulated by neurons and astrocytes. Knowledge of how these cells control blood flow is crucial for understanding how neural computation is powered, for interpreting functional imaging scans of brains, and for developing treatments for neurological disorders. It is now recognized that neurotransmitter-mediated signalling has a key role in regulating cerebral blood flow, that much of this control is mediated by astrocytes, that oxygen modulates blood flow regulation, and that blood flow may be controlled by capillaries as well as by arterioles. These conceptual shifts in our understanding of cerebral blood flow control have important implications for the development of new therapeutic approaches.


Asunto(s)
Encéfalo/irrigación sanguínea , Circulación Cerebrovascular/fisiología , Neuroglía/fisiología , Neuronas/fisiología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/fisiopatología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Isquemia Encefálica/fisiopatología , Humanos , Neurotransmisores/metabolismo , Óxido Nítrico/metabolismo , Oxígeno/metabolismo , Transducción de Señal
8.
J Neurosci ; 34(34): 11504-13, 2014 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-25143628

RESUMEN

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.


Asunto(s)
Microvasos/fisiología , Flujo Sanguíneo Regional/fisiología , Retina/anatomía & histología , Vasos Retinianos/fisiología , Acetamidas/metabolismo , Actinas/metabolismo , Análisis de Varianza , Animales , Antígenos/metabolismo , Dextranos/metabolismo , Eritrocitos/fisiología , Fusión de Flicker , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/metabolismo , Lectinas/metabolismo , Masculino , Microscopía Confocal , Proteoglicanos/metabolismo , Ratas , Ratas Sprague-Dawley , Vasos Retinianos/citología , Factores de Tiempo
9.
J Physiol ; 592(3): 491-504, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24277867

RESUMEN

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.


Asunto(s)
Agonistas del Receptor Purinérgico P2X/farmacología , Antagonistas del Receptor Purinérgico P2X/farmacología , Arteria Retiniana/metabolismo , Vasodilatación , Adenosina Trifosfato/metabolismo , Animales , Apirasa/farmacología , Arteriolas/efectos de los fármacos , Arteriolas/metabolismo , Arteriolas/fisiología , Masculino , Ratas , Ratas Long-Evans , Arteria Retiniana/efectos de los fármacos , Arteria Retiniana/fisiología
10.
Proc Natl Acad Sci U S A ; 108(43): 17827-31, 2011 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-22006332

RESUMEN

Neurovascular coupling is a process through which neuronal activity leads to local increases in blood flow in the central nervous system. In brain slices, 100% O(2) has been shown to alter neurovascular coupling, suppressing activity-dependent vasodilation. However, in vivo, hyperoxia reportedly has no effect on blood flow. Resolving these conflicting findings is important, given that hyperoxia is often used in the clinic in the treatment of both adults and neonates, and a reduction in neurovascular coupling could deprive active neurons of adequate nutrients. Here we address this issue by examining neurovascular coupling in both ex vivo and in vivo rat retina preparations. In the ex vivo retina, 100% O(2) reduced light-evoked arteriole vasodilations by 3.9-fold and increased vasoconstrictions by 2.6-fold. In vivo, however, hyperoxia had no effect on light-evoked arteriole dilations or blood velocity. Oxygen electrode measurements showed that 100% O(2) raised pO(2) in the ex vivo retina from 34 to 548 mm Hg, whereas hyperoxia has been reported to increase retinal pO(2) in vivo to only ~53 mm Hg [Yu DY, Cringle SJ, Alder VA, Su EN (1994) Am J Physiol 267:H2498-H2507]. Replicating the hyperoxic in vivo pO(2) of 53 mm Hg in the ex vivo retina did not alter vasomotor responses, indicating that although O(2) can modulate neurovascular coupling when raised sufficiently high, the hyperoxia-induced rise in retinal pO(2) in vivo is not sufficient to produce a modulatory effect. Our findings demonstrate that hyperoxia does not alter neurovascular coupling in vivo, ensuring that active neurons receive an adequate supply of nutrients.


Asunto(s)
Hipoxia de la Célula/fisiología , Oxígeno/metabolismo , Neuronas Retinianas/metabolismo , Vasos Retinianos/metabolismo , Vasoconstricción/fisiología , Vasodilatación/fisiología , Animales , Masculino , Microelectrodos , Presión Parcial , Estimulación Luminosa , Ratas , Ratas Long-Evans , Transducción de Señal/fisiología , Estadísticas no Paramétricas
11.
J Cereb Blood Flow Metab ; 44(2): 155-168, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-37728791

RESUMEN

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.


Asunto(s)
Hipoglucemia , Insulinas , Acoplamiento Neurovascular , Ratones , Animales , Acoplamiento Neurovascular/fisiología , Arteriolas/metabolismo , Capilares/metabolismo , Circulación Cerebrovascular/fisiología , Vibrisas/fisiología , Hipoglucemia/inducido químicamente , Hipoglucemia/metabolismo , Glucosa/metabolismo , Insulinas/metabolismo , Insulinas/farmacología
12.
Artículo en Inglés | MEDLINE | ID: mdl-38316553

RESUMEN

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.


Asunto(s)
Astrocitos , Circulación Cerebrovascular , Astrocitos/metabolismo , Circulación Cerebrovascular/fisiología , Neuronas , Prostaglandinas/metabolismo
13.
J Undergrad Neurosci Educ ; 12(1): A11-7, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24319385

RESUMEN

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.

14.
J Cereb Blood Flow Metab ; 42(8): 1534-1546, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35296178

RESUMEN

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.


Asunto(s)
Hipoglucemia , Insulinas , Animales , Arteriolas/metabolismo , Astrocitos/metabolismo , Circulación Cerebrovascular/fisiología , Hipoglucemia/metabolismo , Insulinas/metabolismo , Insulinas/farmacología , Ratones , Vasodilatadores/farmacología
15.
Exp Eye Res ; 93(5): 767-70, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21884697

RESUMEN

A micro-advancer device that positions a narrow-gauge needle within the vitreous humor of the rat eye is described. The device is compact, simple and inexpensive to manufacture. It consists of an outer guard needle and an inner injection needle that is advanced through the guard needle. With the rat held in a stereotaxic holder and the globe fixed to a stabilizing ring, the outer 25-gauge guard needle is advanced through the sclera using a standard micromanipulator. The inner 31-gauge injection needle is then advanced through the guard needle with a manually controlled leadscrew and carriage mechanism. The inner injection needle is attached to a Hamilton syringe and can be positioned to within microns of the retinal surface under visual observation through a microscope. The injection needle is fixed to the device by a quick-release clamp on the carriage and can be rapidly exchanged while the guard needle remains in place in the vitreous. This permits different solutions to be injected sequentially into the vitreous humor. Recording electrodes, stimulating electrodes, and optical fibers can also be advanced through the guard needle and positioned accurately near the retinal surface or within the retina.


Asunto(s)
Estimulación Eléctrica/instrumentación , Inyecciones Intravítreas/instrumentación , Retina/fisiología , Cuerpo Vítreo/efectos de los fármacos , Animales , Diseño de Equipo , Microelectrodos , Agujas , Soluciones Oftálmicas/administración & dosificación , Preparaciones Farmacéuticas/administración & dosificación , Ratas
16.
J Neurosci ; 29(36): 11237-45, 2009 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-19741130

RESUMEN

Adenosine is a neuromodulator that activates presynaptic receptors to regulate synaptic transmission and postsynaptic receptors to hyperpolarize neurons. Here, we report that adenosine-induced hyperpolarization of retinal ganglion cells is produced by the activation of A(1) receptors, which initiates a signaling cascade that activates G-protein-coupled inwardly rectifying K(+) (GIRK) channels and small conductance Ca(2+)-activated K(+) (SK) channels. Rat retinal ganglion cells were stimulated by focal ejection of the adenosine receptor agonist 5'-N-ethylcarboxamidoadenosine (NECA) while cell activity was monitored with whole-cell patch recordings and Ca(2+) imaging. Focal ejections of NECA evoked outward currents in all cells tested and reduced light- and depolarization-induced spiking. The NECA-evoked current was abolished by the A(1) antagonist 1,3-dipropyl-8-cyclopentylxanthine (DPCPX) but was unaffected by A(2a), A(2b), and A(3) antagonists, indicating that the response was mediated entirely by A(1) receptors. The GIRK channel blocker rTertiapin-Q diminished the NECA-evoked inhibitory current by 56 +/- 12%, whereas the SK channel blocker apamin decreased the NECA-induced current by 42 +/- 7%. The SK component of the NECA-evoked current coincided with an increase in intracellular Ca(2+) and was blocked by IP(3) receptor antagonists and depletion of internal Ca(2+) stores, suggesting that A(1) receptor activation leads to an increase in IP(3), which then elevates intracellular Ca(2+) and activates SK channels. This A(1)-mediated, prolonged SK channel activation has not been described previously. The coactivation of GIRK and SK channels represents a novel mechanism of adenosine-mediated neuromodulation that could contribute to the regulation of retinal ganglion cell activity.


Asunto(s)
Adenosina/fisiología , Canales de Potasio de Rectificación Interna/fisiología , Receptor de Adenosina A1/fisiología , Receptores Acoplados a Proteínas G/fisiología , Células Ganglionares de la Retina/fisiología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/fisiología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Adenosina/farmacología , Agonistas del Receptor de Adenosina A1 , Adenosina-5'-(N-etilcarboxamida)/farmacología , Animales , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Ratas , Ratas Long-Evans , Células Ganglionares de la Retina/efectos de los fármacos
17.
J Neurosci ; 29(36): 11339-46, 2009 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-19741140

RESUMEN

Intercellular glial Ca(2+) waves constitute a signaling pathway between glial cells. Artificial stimuli have previously been used to evoke these waves, and their physiological significance has been questioned. We report here that Ca(2+) waves occur spontaneously in rat retinal glial cells, both in the isolated retina and in vivo. These spontaneous waves are propagated by ATP release. In the isolated retina, suramin (P2 receptor antagonist) reduces the frequency of spontaneous wave generation by 53%, and apyrase (ATP-hydrolyzing enzyme) reduces frequency by 95-100%. Luciferin-luciferase chemiluminescence reveals waves of ATP matching the spontaneous Ca(2+) waves, indicating that ATP release occurs as spontaneous Ca(2+) waves are generated. Wave generation also depends on age. Spontaneous wave frequency rises from 0.27 to 1.0 per minute per mm(2), as rats age from 20 to 120 d. The sensitivity of glia to ATP does not increase with age, but the ATP released by evoked waves is 31% greater in 120-d-old than in 20-d-old rats, suggesting that increased ATP release in older animals could account for the higher frequency of wave generation. Simultaneous imaging of glial Ca(2+) and arterioles in the isolated retina demonstrates that spontaneous waves alter vessel diameter, implying that spontaneous waves may have a significant impact on retinal physiology. Spontaneous intercellular glial Ca(2+) waves also occur in the retina in vivo, with frequency, speed, and diameter similar to the isolated retina. Increased spontaneous wave occurrence with age suggests that wave generation may be related to retinal pathology.


Asunto(s)
Envejecimiento/fisiología , Señalización del Calcio/fisiología , Neuroglía/fisiología , Retina/crecimiento & desarrollo , Factores de Edad , Animales , Masculino , Ratas , Ratas Long-Evans , Transmisión Sináptica/fisiología
18.
Glia ; 58(16): 1996-2004, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20830810

RESUMEN

Neuronal activity leads to arteriole dilation and increased blood flow in retinal vessels. This response, termed functional hyperemia, is diminished in the retinas of diabetic patients, possibly contributing to the development of diabetic retinopathy. The mechanism responsible for this loss is unknown. Here we show that light-evoked arteriole dilation was reduced by 58% in a streptozotocin-induced rat model of type 1 diabetes. Functional hyperemia is believed to be mediated by glial cells and we found that glial-evoked vasodilation was reduced by 60% in diabetic animals. The diabetic retinas showed neither a decrease in the thickness of the retinal layers nor an increase in neuronal loss, although signs of early glial reactivity and an upregulation of inducible nitric oxide synthase (iNOS) were detected. Inhibition of iNOS restored both light- and glial-evoked dilations to control levels. These findings suggest that high NO levels resulting from iNOS upregulation alters glial control of vessel diameter and may underlie the loss of functional hyperemia observed in diabetic retinopathy. Restoring functional hyperemia by iNOS inhibition may limit the progression of retinopathy in diabetic patients.


Asunto(s)
Retinopatía Diabética/tratamiento farmacológico , Guanidinas/farmacología , Hiperemia/tratamiento farmacológico , Neuroglía/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Retina/efectos de los fármacos , Animales , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Retinopatía Diabética/metabolismo , Retinopatía Diabética/patología , Inhibidores Enzimáticos/farmacología , Hiperemia/metabolismo , Hiperemia/patología , Masculino , Neuroglía/metabolismo , Neuroglía/patología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Estimulación Luminosa , Ratas , Ratas Long-Evans , Retina/enzimología , Retina/patología , Vasos Retinianos/efectos de los fármacos , Vasos Retinianos/fisiología , Vasodilatación/efectos de los fármacos , Vasodilatación/fisiología
19.
J Neurophysiol ; 103(3): 1357-74, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20053849

RESUMEN

Trains of action potentials of rat and cat retinal ganglion cells (RGCs) were recorded intracellularly across a temperature range of 7-37 degrees C. Phase plots of the experimental impulse trains were precision fit using multicompartment simulations of anatomically reconstructed rat and cat RGCs. Action potential excitation was simulated with a "Five-channel model" [Na, K(delayed rectifier), Ca, K(A), and K(Ca-activated) channels] and the nonspace-clamped condition of the whole cell recording was exploited to determine the channels' distribution on the dendrites, soma, and proximal axon. At each temperature, optimal phase-plot fits for RGCs occurred with the same unique channel distribution. The "waveform" of the electrotonic current was found to be temperature dependent, which reflected the shape changes in the experimental action potentials and confirmed the channel distributions. The distributions are cell-type specific and adequate for soma and dendritic excitation with a safety margin. The highest Na-channel density was found on an axonal segment some 50-130 microm distal to the soma, as determined from the temperature-dependent "initial segment-somadendritic (IS-SD) break." The voltage dependence of the gating rate constants remains invariant between 7 and 23 degrees C and between 30 and 37 degrees C, but undergoes a transition between 23 and 30 degrees C. Both gating-kinetic and ion-permeability Q10s remain virtually constant between 23 and 37 degrees C (kinetic Q10s = 1.9-1.95; permeability Q10s = 1.49-1.64). The Q10s systematically increase for T <23 degrees C (kinetic Q10 = 8 at T = 8 degrees C). The Na channels were consistently "sleepy" (non-Arrhenius) for T <8 degrees C, with a loss of spiking for T <7 degrees C.


Asunto(s)
Canales Iónicos/fisiología , Células Ganglionares de la Retina/fisiología , Potenciales de Acción/fisiología , Algoritmos , Animales , Gatos , Citoplasma/metabolismo , Interpretación Estadística de Datos , Estimulación Eléctrica , Sinapsis Eléctricas/fisiología , Electrofisiología , Técnicas In Vitro , Activación del Canal Iónico/fisiología , Cinética , Modelos Neurológicos , Técnicas de Placa-Clamp , Ratas , Ratas Long-Evans , Células Ganglionares de la Retina/ultraestructura , Temperatura
20.
J Neurosurg ; 132(6): 1820-1828, 2019 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-30952117

RESUMEN

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.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA