Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Physiol Rev ; 96(3): 911-73, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27252279

RESUMEN

Transient receptor potential vanilloid type 4 (TRPV4) is a calcium-permeable nonselective cation channel, originally described in 2000 by research teams led by Schultz (Nat Cell Biol 2: 695-702, 2000) and Liedtke (Cell 103: 525-535, 2000). TRPV4 is now recognized as being a polymodal ionotropic receptor that is activated by a disparate array of stimuli, ranging from hypotonicity to heat and acidic pH. Importantly, this ion channel is constitutively expressed and capable of spontaneous activity in the absence of agonist stimulation, which suggests that it serves important physiological functions, as does its widespread dissemination throughout the body and its capacity to interact with other proteins. Not surprisingly, therefore, it has emerged more recently that TRPV4 fulfills a great number of important physiological roles and that various disease states are attributable to the absence, or abnormal functioning, of this ion channel. Here, we review the known characteristics of this ion channel's structure, localization and function, including its activators, and examine its functional importance in health and disease.


Asunto(s)
Canalopatías/metabolismo , Canales Catiónicos TRPV/fisiología , Animales , Canalopatías/genética , Humanos , Ratones
2.
Curr Top Membr ; 85: 187-226, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32402639

RESUMEN

Retinal pressure autoregulation is an important mechanism that protects the retina by stabilizing retinal blood flow during changes in arterial or intraocular pressure. Similar to other vascular beds, retinal pressure autoregulation is thought to be mediated largely through the myogenic response of small arteries and arterioles which constrict when transmural pressure increases or dilate when it decreases. Over recent years, we and others have investigated the signaling pathways underlying the myogenic response in retinal arterioles, with particular emphasis on the involvement of different ion channels expressed in the smooth muscle layer of these vessels. Here, we review and extend previous work on the expression and spatial distribution of the plasma membrane and sarcoplasmic reticulum ion channels present in retinal vascular smooth muscle cells (VSMCs) and discuss their contribution to pressure-induced myogenic tone in retinal arterioles. This includes new data demonstrating that several key players and modulators of the myogenic response show distinctively heterogeneous expression along the length of the retinal arteriolar network, suggesting differences in myogenic signaling between larger and smaller pre-capillary arterioles. Our immunohistochemical investigations have also highlighted the presence of actin-containing microstructures called myobridges that connect the retinal VSMCs to one another. Although further work is still needed, studies to date investigating myogenic mechanisms in the retina have contributed to a better understanding of how blood flow is regulated in this tissue. They also provide a basis to direct future research into retinal diseases where blood flow changes contribute to the pathology.


Asunto(s)
Arteriolas/fisiología , Canales Iónicos/metabolismo , Desarrollo de Músculos , Retina/fisiología , Animales , Arteriolas/metabolismo , Fenómenos Biomecánicos , Homeostasis , Humanos
3.
Diabetologia ; 61(12): 2654-2667, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30112688

RESUMEN

AIMS/HYPOTHESIS: Recent studies suggest that abnormal function in Müller glial cells plays an important role in the pathogenesis of diabetic retinopathy. This is associated with the selective accumulation of the acrolein-derived advanced lipoxidation end-product, Nε-(3-formyl-3,4-dehydropiperidino)lysine (FDP-lysine), on Müller cell proteins. The aim of the current study was to identify more efficacious acrolein-scavenging drugs and determine the effects of the most potent on Müller cell FDP-lysine accumulation and neuroretinal dysfunction during diabetes. METHODS: An ELISA-based in vitro assay was optimised to compare the acrolein-scavenging abilities of a range of drugs. This identified 2-hydrazino-4,6-dimethylpyrimidine (2-HDP) as a new and potent acrolein scavenger. The ability of this agent to modify the development of diabetic retinopathy was tested in vivo. Male Sprague Dawley rats were divided into three groups: (1) non-diabetic; (2) streptozotocin-induced diabetic; and (3) diabetic treated with 2-HDP in their drinking water for the duration of diabetes. Liquid chromatography high-resolution mass spectrometry was used to detect 2-HDP reaction products in the retina. Immunohistochemistry, real-time quantitative (q)RT-PCR and electroretinography were used to assess retinal changes 3 months after diabetes induction. RESULTS: 2-HDP was the most potent of six acrolein-scavenging agents tested in vitro (p < 0.05). In vivo, administration of 2-HDP reduced Müller cell accumulation of FDP-lysine at 3 months in rats rendered diabetic with streptozotocin (p < 0.001). A 2-HDP adduct was identified in the retinas of diabetic animals treated with this compound. 2-HDP supplementation was associated with reduced Müller cell gliosis (p < 0.05), reduced expression of the oxidative stress marker haem oxygenase-1 (p < 0.001) and partial normalisation of inwardly rectifying K+ channel 4.1 (Kir4.1) expression (p < 0.001 for staining in perivascular regions and the innermost region of the ganglion cell layer). Diabetes-induced retinal expression of inflammatory markers, inflammatory signalling compounds and activation of retinal microglial cells were all reduced in 2-HDP-treated animals. Retinal neurophysiological defects in diabetic animals, as indicated by changes in the electroretinogram 7 weeks after induction of diabetes, were also reduced by 2-HDP (p < 0.05-0.01 for b-wave amplitudes at flash intensities from -10 to +10 dB; p < 0.01 for time to peak of summed oscillatory potentials at +10 dB). CONCLUSIONS/INTERPRETATION: These findings support the hypothesis that Müller cell accumulation of FDP-lysine plays an important role in the development of diabetic retinopathy. Our results also suggest that 2-HDP may have therapeutic potential for delaying or treating this sight-threatening complication.


Asunto(s)
Acroleína/toxicidad , Células Ependimogliales/efectos de los fármacos , Células Ependimogliales/metabolismo , Depuradores de Radicales Libres/uso terapéutico , Lisina/metabolismo , Animales , Supervivencia Celular/efectos de los fármacos , Cromatografía Liquida , Retinopatía Diabética/metabolismo , Electrorretinografía , Ensayo de Inmunoadsorción Enzimática , Inmunohistoquímica , Masculino , Espectrometría de Masas , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
4.
Eur Respir J ; 50(3)2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28931666

RESUMEN

The cough reflex becomes hyperresponsive in acute and chronic respiratory diseases, but understanding the underlying mechanism is hampered by difficulty accessing human tissue containing both nerve endings and neuronal cell bodies. We refined an adult stem cell sensory neuronal model to overcome the limited availability of human neurones and applied the model to study transient receptor potential ankyrin 1 (TRPA1) channel expression and activation.Human dental pulp stem cells (hDPSCs) were differentiated towards a neuronal phenotype, termed peripheral neuronal equivalents (PNEs). Using molecular and immunohistochemical techniques, together with Ca2+ microfluorimetry and whole cell patch clamping, we investigated roles for nerve growth factor (NGF) and the viral mimic poly I:C in TRPA1 activation.PNEs exhibited morphological, molecular and functional characteristics of sensory neurons and expressed functional TRPA1 channels. PNE treatment with NGF for 20 min generated significantly larger inward and outward currents compared to untreated PNEs in response to the TRPA1 agonist cinnamaldehyde (p<0.05). PNE treatment with poly I:C caused similar transient heightened responses to TRPA1 activation compared to untreated cells.Using the PNE neuronal model we observed both NGF and poly I:C mediated sensory neuronal hyperresponsiveness, representing potential neuro-inflammatory mechanisms associated with heightened nociceptive responses recognised in cough hypersensitivity syndrome.


Asunto(s)
Tos/fisiopatología , Factor de Crecimiento Nervioso/farmacología , Proteínas del Tejido Nervioso/metabolismo , Canal Catiónico TRPA1/metabolismo , Canales de Calcio/metabolismo , Tos/tratamiento farmacológico , Pulpa Dental/citología , Humanos , Neuronas Aferentes/citología , Poli I-C/farmacología , Células Madre/efectos de los fármacos , Canal Catiónico TRPA1/genética , Canales Catiónicos TRPV/metabolismo
5.
Exp Eye Res ; 120: 15-9, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24333760

RESUMEN

Simultaneous non-invasive visualization of blood vessels and nerves in patients can be obtained in the eye. The retinal vasculature is a target of many retinopathies. Inflammation, readily manifest by leukocyte adhesion to the endothelial lining, is a key pathophysiological mechanism of many retinopathies, making it a valuable and ubiquitous target for disease research. Leukocyte fluorography has been extensively used in the past twenty years; however, fluorescent markers, visualization techniques, and recording methods have differed between studies. The lack of detailed protocol papers regarding leukocyte fluorography, coupled with lack of uniformity between studies, has led to a paucity of standards for leukocyte transit (velocity, adherence, extravasation) in the retina. Here, we give a detailed description of a convenient method using acridine orange (AO) and a commercially available scanning laser ophthalmoscope (SLO, HRA-OCT Spectralis) to view leukocyte behavior in the mouse retina. Normal mice are compared to mice with acute and chronic inflammation. This method can be readily adopted in many research labs.


Asunto(s)
Naranja de Acridina , Angiografía con Fluoresceína , Colorantes Fluorescentes , Leucocitos/fisiología , Arteria Retiniana/fisiología , Vena Retiniana/fisiología , Animales , Velocidad del Flujo Sanguíneo , Movimiento Celular/fisiología , Diabetes Mellitus Tipo 1/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Microscopía Confocal , Oftalmoscopios , Flujo Sanguíneo Regional/fisiología , Vasculitis Retiniana/inducido químicamente , Vasculitis Retiniana/fisiopatología , Tomografía de Coherencia Óptica , Factor A de Crecimiento Endotelial Vascular/farmacología , Grabación en Video
6.
J Biol Chem ; 287(5): 2963-70, 2012 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-22128172

RESUMEN

Transient receptor potential (TRP) channels couple various environmental factors to changes in membrane potential, calcium influx, and cell signaling. They also integrate multiple stimuli through their typically polymodal activation. Thus, although the TRPM8 channel has been extensively investigated as the major neuronal cold sensor, it is also regulated by various chemicals, as well as by several short channel isoforms. Mechanistic understanding of such complex regulation is facilitated by quantitative single-channel analysis. We have recently proposed a single-channel mechanism of TRPM8 regulation by voltage and temperature. Using this gating mechanism, we now investigate TRPM8 inhibition in cell-attached patches using HEK293 cells expressing TRPM8 alone or coexpressed with its short sM8-6 isoform. This is compared with inhibition by the chemicals N-(4-tert-butylphenyl)-4-(3-chloropyridin-2-yl)piperazine-1-carboxamide (BCTC) and clotrimazole or by elevated temperature. We found that within the seven-state single-channel gating mechanism, inhibition of TRPM8 by short sM8-6 isoforms closely resembles inhibition by increased temperature. In contrast, inhibition by BCTC and that by clotrimazole share a different set of common features.


Asunto(s)
Antifúngicos/farmacología , Clotrimazol/farmacología , Calor , Activación del Canal Iónico/efectos de los fármacos , Activación del Canal Iónico/fisiología , Pirazinas/farmacología , Piridinas/farmacología , Canales Catiónicos TRPM/antagonistas & inhibidores , Canales Catiónicos TRPM/metabolismo , Células HEK293 , Humanos , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Canales Catiónicos TRPM/genética , Termorreceptores/metabolismo
7.
JCI Insight ; 7(18)2022 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-36134661

RESUMEN

Loss of retinal blood flow autoregulation is an early feature of diabetes that precedes the development of clinically recognizable diabetic retinopathy (DR). Retinal blood flow autoregulation is mediated by the myogenic response of the retinal arterial vessels, a process that is initiated by the stretch­dependent activation of TRPV2 channels on the retinal vascular smooth muscle cells (VSMCs). Here, we show that the impaired myogenic reaction of retinal arterioles from diabetic animals is associated with a complete loss of stretch­dependent TRPV2 current activity on the retinal VSMCs. This effect could be attributed, in part, to TRPV2 channel downregulation, a phenomenon that was also evident in human retinal VSMCs from diabetic donors. We also demonstrate that TRPV2 heterozygous rats, a nondiabetic model of impaired myogenic reactivity and blood flow autoregulation in the retina, develop a range of microvascular, glial, and neuronal lesions resembling those observed in DR, including neovascular complexes. No overt kidney pathology was observed in these animals. Our data suggest that TRPV2 dysfunction underlies the loss of retinal blood flow autoregulation in diabetes and provide strong support for the hypothesis that autoregulatory deficits are involved in the pathogenesis of DR.


Asunto(s)
Diabetes Mellitus , Retinopatía Diabética , Arteria Retiniana , Animales , Arteriolas , Homeostasis/fisiología , Humanos , Ratas , Vasos Retinianos , Canales Catiónicos TRPV/genética
8.
Am J Physiol Cell Physiol ; 301(3): C717-28, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21633080

RESUMEN

Studies concerning the physiological significance of Ca(2+) sparks often depend on the detection and measurement of large populations of events in noisy microscopy images. Automated detection methods have been developed to quickly and objectively distinguish potential sparks from noise artifacts. However, previously described algorithms are not suited to the reliable detection of sparks in images where the local baseline fluorescence and noise properties can vary significantly, and risk introducing additional bias when applied to such data sets. Here, we describe a new, conceptually straightforward approach to spark detection in linescans that addresses this issue by combining variance stabilization with local baseline subtraction. We also show that in addition to greatly increasing the range of images in which sparks can be automatically detected, the use of a more accurate noise model enables our algorithm to achieve similar detection sensitivities with fewer false positives than previous approaches when applied both to synthetic and experimental data sets. We propose, therefore, that it might be a useful tool for improving the reliability and objectivity of spark analysis in general, and describe how it might be further optimized for specific applications.


Asunto(s)
Algoritmos , Señalización del Calcio/fisiología , Procesamiento de Imagen Asistido por Computador/métodos , Animales , Arteriolas/metabolismo , Simulación por Computador , Microscopía Confocal/métodos , Microscopía Fluorescente/métodos , Músculo Liso Vascular/metabolismo , Valor Predictivo de las Pruebas , Ratas , Ratas Sprague-Dawley , Arteria Retiniana/metabolismo , Relación Señal-Ruido
9.
Exp Physiol ; 95(11): 1049-60, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20696785

RESUMEN

Ever since it was shown that maintenance of muscle contraction required the presence of extracellular Ca(2+), evidence has accumulated that Ca(2+) plays a crucial role in excitation-contraction coupling. This culminated in the use of the photoprotein aequorin to demonstrate that [Ca(2+)](i) increased after depolarization but before contraction in barnacle muscle. Green fluorescent protein was extracted from the same jellyfish as aequorin, so this work also has important historical links to the use of fluorescent proteins as markers in living cells. The subsequent development of cell-permeant Ca(2+) indicators resulted in a dramatic increase in related research, revealing Ca(2+) to be a ubiquitous cell signal. High-speed, confocal Ca(2+) imaging has now revealed subcellular detail not previously apparent, with the identification of Ca(2+) sparks. These act as building blocks for larger transients during excitation-contraction coupling in cardiac muscle, but their function in smooth muscle appears more diverse, with evidence suggesting both 'excitatory' and 'inhibitory' roles. Sparks can activate Ca(2+)-sensitive Cl() and K(+) currents, which exert positive and negative feedback, respectively, on global Ca(2+) signalling, through changes in membrane potential and activation of voltage-operated Ca(2+) channels. Calcium imaging has also demonstrated that agonists that appear to evoke relatively tonic increases in average [Ca(2+)](i) at the whole tissue level often stimulate much higher frequency phasic Ca(2+) oscillations at the cellular level. These findings may require re-evaluation of some of our models of Ca(2+) signalling to account for newly revealed cellular and subcellular detail. Future research in the field is likely to make increasing use of genetically coded Ca(2+) indicators expressed in an organelle- or tissue-specific manner.


Asunto(s)
Señalización del Calcio/fisiología , Calcio/metabolismo , Animales , Canales de Calcio/metabolismo , Soluciones Isotónicas/farmacología , Contracción Muscular/efectos de los fármacos , Contracción Muscular/fisiología , Solución de Ringer
10.
Circ Res ; 100(5): 703-11, 2007 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-17293477

RESUMEN

Retinal vasoconstriction and reduced retinal blood flow precede the onset of diabetic retinopathy. The pathophysiological mechanisms that underlie increased retinal arteriolar tone during diabetes remain unclear. Normally, local Ca(2+) release events (Ca(2+)-sparks), trigger the activation of large-conductance Ca(2+)-activated K(+)(BK)-channels which hyperpolarize and relax vascular smooth muscle cells, thereby causing vasodilatation. In the present study, we examined BK channel function in retinal vascular smooth muscle cells from streptozotocin-induced diabetic rats. The BK channel inhibitor, Penitrem A, constricted nondiabetic retinal arterioles (pressurized to 70mmHg) by 28%. The BK current evoked by caffeine was dramatically reduced in retinal arterioles from diabetic animals even though caffeine-evoked [Ca(2+)](i) release was unaffected. Spontaneous BK currents were smaller in diabetic cells, but the amplitude of Ca(2+)-sparks was larger. The amplitudes of BK currents elicited by depolarizing voltage steps were similar in control and diabetic arterioles and mRNA expression of the pore-forming BKalpha subunit was unchanged. The Ca(2+)-sensitivity of single BK channels from diabetic retinal vascular smooth muscle cells was markedly reduced. The BKbeta1 subunit confers Ca(2+)-sensitivity to BK channel complexes and both transcript and protein levels for BKbeta1 were appreciably lower in diabetic retinal arterioles. The mean open times and the sensitivity of BK channels to tamoxifen were decreased in diabetic cells, consistent with a downregulation of BKbeta1 subunits. The potency of blockade by Pen A was lower for BK channels from diabetic animals. Thus, changes in the molecular composition of BK channels could account for retinal hypoperfusion in early diabetes, an idea having wider implications for the pathogenesis of diabetic hypertension.


Asunto(s)
Calcio/fisiología , Diabetes Mellitus Experimental/metabolismo , Regulación hacia Abajo/fisiología , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/antagonistas & inhibidores , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/biosíntesis , Músculo Liso Vascular/metabolismo , Arteria Retiniana/metabolismo , Animales , Arteriolas/metabolismo , Subunidades beta de los Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Ratas , Transcripción Genética
11.
Invest Ophthalmol Vis Sci ; 60(10): 3297-3309, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31369032

RESUMEN

Purpose: We investigate the contribution of TRPV1 and TRPV4 channels to retinal angiogenesis. Methods: Primary retinal microvascular endothelial cells (RMECs) were used for RT-PCR, Western blotting, immunolabeling, Ca2+ signaling, and whole-cell patch-clamp studies while localization of TRPV1 also was assessed in retinal endothelial cells using whole mount preparations. The effects of pharmacologic blockers of TRPV1 and TRPV4 on retinal angiogenic activity was evaluated in vitro using sprout formation, cell migration, proliferation, and tubulogenesis assays, and in vivo using the mouse model of oxygen-induced retinopathy (OIR). Heteromultimerization of TRPV1 and TRPV4 channels in RMECs was assessed using proximity ligation assays (PLA) and electrophysiologic recording. Results: TRPV1 mRNA and protein expression were identified in RMECs. TRPV1 labelling was found to be mainly localized to the cytoplasm with some areas of staining colocalizing with the plasma membrane. Staining patterns for TRPV1 were broadly similar in endothelial cells of intact vessels within retinal flat mounts. Functional expression of TRPV1 and TRPV4 in RMECs was confirmed by patch-clamp recording. Pharmacologic inhibition of TRPV1 or TRPV4 channels suppressed in vitro retinal angiogenesis through a mechanism involving the modulation of tubulogenesis. Blockade of these channels had no effect on VEGF-stimulated angiogenesis or Ca2+ signals in vitro. PLA and patch-clamp studies revealed that TRPV1 and TRPV4 form functional heteromeric channel complexes in RMECs. Inhibition of either channel reduced retinal neovascularization and promoted physiologic revascularization of the ischemic retina in the OIR mouse model. Conclusions: TRPV1 and TRPV4 channels represent promising targets for therapeutic intervention in vasoproliferative diseases of the retina.


Asunto(s)
Células Endoteliales/metabolismo , Neovascularización Retiniana/metabolismo , Vasos Retinianos/citología , Canales Catiónicos TRPV/fisiología , Animales , Animales Recién Nacidos , Western Blotting , Calcio/metabolismo , Señalización del Calcio/fisiología , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Oxígeno/toxicidad , Técnicas de Placa-Clamp , Piridinas/farmacología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Neovascularización Retiniana/patología , Sulfonamidas/farmacología , Sulfonas/farmacología , Canales Catiónicos TRPV/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/farmacología
12.
JCI Insight ; 4(6)2019 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-30721154

RESUMEN

While anti-VEGF drugs are commonly used to inhibit pathological retinal and choroidal neovascularization, not all patients respond in an optimal manner. Mechanisms underpinning resistance to anti­VEGF therapy include the upregulation of other proangiogenic factors. Therefore, therapeutic strategies that simultaneously target multiple growth factor signaling pathways would have significant value. Here, we show that Ca2+/calmodulin-dependent kinase II (CAMKII) mediates the angiogenic actions of a range of growth factors in human retinal endothelial cells and that this kinase acts as a key nodal point for the activation of several signal transduction cascades that are known to play a critical role in growth factor-induced angiogenesis. We also demonstrate that endothelial CAMKIIγ and -δ isoforms differentially regulate the angiogenic effects of different growth factors and that genetic deletion of these isoforms suppresses pathological retinal and choroidal neovascularization in vivo. Our studies suggest that CAMKII could provide a novel and efficacious target to inhibit multiple angiogenic signaling pathways for the treatment of vasoproliferative diseases of the eye. CAMKIIγ represents a particularly promising target, as deletion of this isoform inhibited pathological neovascularization, while enhancing reparative angiogenesis in the ischemic retina.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/efectos de los fármacos , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Neovascularización Coroidal/tratamiento farmacológico , Retina/efectos de los fármacos , Inductores de la Angiogénesis/farmacología , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Supervivencia Celular/efectos de los fármacos , Neovascularización Coroidal/patología , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Cinetina/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Isoformas de Proteínas , Proteómica , Retina/patología , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular
13.
Cell Calcium ; 43(2): 142-54, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17574671

RESUMEN

Spontaneous Ca(2+)-sparks were imaged using confocal line scans of fluo-4 loaded myocytes in retinal arterioles. Tetracaine produced concentration-dependent decreases in spark frequency, and modified the spatiotemporal characteristics of residual sparks. Tetracaine (10 microM) reduced the rate of rise but prolonged the average rise time so that average spark amplitude was unaltered. The mean half-time of spark decay was also unaffected, suggesting that spark termination, although delayed, remained well synchronized. Sparks spread transversely across the myocytes in these vessels, and the speed of spread within individual sparks was slowed by approximately 60% in 10 microM tetracaine, as expected if the spark was propagated across the cell but the average P(o) for RyRs was reduced. Staining of isolated vessels with BODIPY-ryanodine and di-4-ANEPPS showed that RyRs were located both peripherally, adjacent to the plasma membrane, and in transverse extensions of the SR from one side of the cell to the other. Immuno-labelling of retinal flat mounts demonstrated the presence RyR(2) in arteriole smooth muscle but not RyR(1). We conclude that Ca(2+)-sparks in smooth muscle can result from sequential activation of RyRs distributed over an area of several microm(2), rather than from tightly clustered channels as in striated muscle.


Asunto(s)
Señalización del Calcio/fisiología , Músculo Liso Vascular/fisiología , Canal Liberador de Calcio Receptor de Rianodina/fisiología , Tetracaína/farmacología , Animales , Cafeína/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Señalización del Calcio/efectos de los fármacos , Masculino , Miocitos del Músculo Liso/fisiología , Ratas , Ratas Sprague-Dawley , Arteria Retiniana , Canal Liberador de Calcio Receptor de Rianodina/efectos de los fármacos
14.
J Vis Exp ; (137)2018 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-30059036

RESUMEN

The retina is a highly metabolically active tissue that requires a substantial blood supply. The retinal circulation supports the inner retina, while the choroidal vessels supply the photoreceptors. Alterations in retinal perfusion contribute to numerous sight-threatening disorders, including diabetic retinopathy, glaucoma and retinal branch vein occlusions. Understanding the molecular mechanisms involved in the control of blood flow through the retina and how these are altered during ocular disease could lead to the identification of new targets for the treatment of these conditions. Retinal arterioles are the main resistance vessels of the retina, and consequently, play a key role in regulating retinal hemodynamics through changes in luminal diameter. In recent years, we have developed methods for isolating arterioles from the rat retina which are suitable for a wide range of applications including cell physiology studies. This preparation has already begun to yield new insights into how blood flow is controlled in the retina and has allowed us to identify some of the key changes that occur during ocular disease. In this article, we describe methods for the isolation of rat retinal arterioles and include protocols for their use in patch-clamp electrophysiology, calcium imaging and pressure myography studies. These vessels are also amenable for use in PCR-, western blotting- and immunohistochemistry-based studies.


Asunto(s)
Arteriolas/fisiología , Fenómenos Fisiológicos Celulares/fisiología , Vasos Retinianos/fisiología , Animales , Humanos , Ratones , Retina
15.
Adv Physiol Educ ; 30(3): 108-12, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16912144

RESUMEN

Capillary filtration is a key area in the understanding of cardiovascular function and has both physiological and pathophysiological relevance in nearly every organ system. This article describes how classic papers in the Legacy collection of American Physiological Society publications can be used in a teaching symposium exploring the evidence supporting current concepts of capillary fluid exchange. Individual students are given papers to read, edit, and present to the class. The appropriate selection and sequencing of these papers allows the development of important physiological concepts to be tracked. A series of papers concerned with capillary filtration is suggested, and the contribution of each to the developing story is outlined. This approach allows students to develop critical and presentation skills and provides them with a case study of the scientific method as it is applied to physiology as well as establishing an appropriate knowledge base concerning the role of hydrostatic and oncotic forces in capillary fluid exchange. Relevant teaching points are explored further using questions based on a figure from one of the three classic papers used: "Microinjection studies of capillary permeability: II. The relationship between capillary pressure and the rate at which fluid passes through the walls of single capillaries," by E. M. Landis (Am J Physiol 82: 217-238, 1927).


Asunto(s)
Permeabilidad Capilar/fisiología , Fisiología/educación , Fisiología/historia , Materiales de Enseñanza , Enseñanza/métodos , Historia del Siglo XX , Humanos
16.
Invest Ophthalmol Vis Sci ; 57(11): 4762-71, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27618414

RESUMEN

PURPOSE: We studied whether the accumulation of advanced lipoxidation end-products (ALEs) in the diabetic retina is linked to the impairment of lipid aldehyde detoxification mechanisms. METHODS: Retinas were collected from nondiabetic and diabetic rats and processed for conventional and quantitative RT-PCR (qRT-PCR), Western blotting, immunohistochemistry, and aldehyde dehydrogenase (ALDH) activity assays. The effect of the ALDH1a1 inhibitor, NCT-501, on ALE accumulation and cell viability in cultured Müller glia also was investigated. RESULTS: The rat retina expressed a range of lipid aldehyde detoxifying ALDH and aldo-keto reductase (AKR) genes. In diabetes, mRNA levels were reduced for 5 of 9 transcripts tested. These findings contrasted with those in the lens and cornea where many of these enzymes were upregulated. We have reported previously accumulation of the acrolein (ACR)-derived ALE, FDP-lysine, in retinal Müller glia during diabetes. In the present study, we show that the main ACR-detoxifying ALDH and AKR genes expressed in the retina, namely, ALDH1a1, ALDH2, and AKR1b1, are principally localized to Müller glia. Diabetes-induced FDP-lysine accumulation in Müller glia was associated with a reduction in ALDH1a1 mRNA and protein expression in whole retina and a decrease in ALDH1a1-immunoreactivity specifically within these cells. No such changes were detected for ALDH2 or AKR1b1. Activity of ALDH was suppressed in the diabetic retina and blockade of ALDH1a1 in cultured Müller glia triggered FDP-lysine accumulation and reduced cell viability. CONCLUSIONS: These findings suggest that downregulation of ALDH and AKR enzymes, particularly ALDH1a1, may contribute ALE accumulation in the diabetic retina.


Asunto(s)
Aldehído Deshidrogenasa/metabolismo , Diabetes Mellitus Experimental , Retinopatía Diabética/metabolismo , Regulación de la Expresión Génica , ARN/genética , Retina/metabolismo , Retinal-Deshidrogenasa/genética , Familia de Aldehído Deshidrogenasa 1 , Animales , Western Blotting , Recuento de Células , Células Cultivadas , Retinopatía Diabética/patología , Células Ependimogliales/metabolismo , Células Ependimogliales/patología , Inmunohistoquímica , Masculino , Microscopía Confocal , Ratas , Ratas Sprague-Dawley , Retina/patología , Retinal-Deshidrogenasa/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
Invest Ophthalmol Vis Sci ; 57(13): 5637-5647, 2016 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-27784066

RESUMEN

PURPOSE: Activation of the transient receptor potential channels, TRPC6, TRPM4, and TRPP1 (PKD2), has been shown to contribute to the myogenic constriction of cerebral arteries. In the present study we sought to determine the potential role of various mechanosensitive TRP channels to myogenic signaling in arterioles of the rat retina. METHODS: Rat retinal arterioles were isolated for RT-PCR, Fura-2 Ca2+ microfluorimetry, patch-clamp electrophysiology, and pressure myography studies. In some experiments, confocal immunolabeling of wholemount preparations was used to examine the localization of specific mechanosensitive TRP channels in retinal vascular smooth muscle cells (VSMCs). RESULTS: Reverse transcription-polymerase chain reaction analysis demonstrated mRNA expression for TRPC1, M7, V1, V2, V4, and P1, but not TRPC6 or M4, in isolated retinal arterioles. Immunolabeling revealed plasma membrane, cytosolic and nuclear expression of TRPC1, M7, V1, V2, V4, and P1 in retinal VSMCs. Hypoosmotic stretch-induced Ca2+ influx in retinal VSMCs was reversed by the TRPV2 inhibitor tranilast and the nonselective TRPP1/V2 antagonist amiloride. Inhibitors of TRPC1, M7, V1, and V4 had no effect. Hypoosmotic stretch-activated cation currents were similar in Na+ and Cs+ containing solutions suggesting no contribution by TRPP1 channels. Direct plasma membrane stretch triggered cation current activity that was blocked by tranilast and specific TRPV2 pore-blocking antibodies and mimicked by the TRPV2 activator, Δ9-tetrahydrocannabinol. Preincubation of retinal arterioles with TRPV2 blocking antibodies prevented the development of myogenic tone. CONCLUSIONS: Our results suggest that retinal VSMCs express a range of mechanosensitive TRP channels, but only TRPV2 appears to contribute to myogenic signaling in this vascular bed.


Asunto(s)
Arteriolas/fisiología , Regulación de la Expresión Génica , Músculo Liso Vascular/fisiología , ARN/genética , Arteria Retiniana/fisiología , Canales Catiónicos TRPV/metabolismo , Vasoconstricción/genética , Animales , Inmunohistoquímica , Masculino , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
18.
Invest Ophthalmol Vis Sci ; 46(9): 3281-7, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16123430

RESUMEN

PURPOSE: By their control of membrane potential and intracellular free Ca(2+) ([Ca(2+)](i)), K(+) currents are pivotal in the regulation of arterial smooth muscle tone. The goal of the present study was to identify and characterize the A-type K(+) current in retinal microvascular smooth muscle (MVSM) and to examine its role in modulating membrane potential and cellular contractility. METHODS: Whole-cell perforated patch-clamp recordings were made from MVSM cells within intact isolated arteriolar segments. Before patch-clamping, retinal arterioles were anchored in the physiological recording bath and perfused with an enzyme cocktail to remove surface basal lamina and to uncouple electrically the endothelial cells from the overlying MVSM cells. RESULTS: K(+) currents were activated by depolarizing steps from -80 to +100 mV in 20-mV increments. A dominant, noninactivating current was elicited by depolarization to potentials positive of -50 mV. Inhibition of this current by 100 nM of the Ca(2+)-activated K(+) channel blocker, Penitrem A, revealed a rapidly inactivating K(+) current that resembled an A-type current. The A-type current was insensitive to tetraethylammonium (TEA) at 1 mM, but was partially suppressed by higher concentrations (10 mM). 4-Aminopyridine (10 mM; 4-AP) completely blocked the A-type current. The 4-AP-sensitive transient current was activated at a potential of -60 mV with peak current densities averaging 29.7 +/- 5.68 pA/pF at +60 mV. The voltage of half-inactivation was -28.3 +/- 1.9 mV, and the time constant for recovery from inactivation at +60 mV was 118.7 +/- 7.9 ms. Under current-clamp conditions 4-AP depolarized the membrane potential by approximately 3 to 4 mV and triggered small contractions and relaxations of individual MVSM cells within the walls of the arterioles. CONCLUSIONS: A-type current is the major voltage-dependent K(+) current in retinal MVSM and appears to play a physiological role in suppressing cell excitability and contractility.


Asunto(s)
Músculo Liso Vascular/fisiología , Canales de Potasio con Entrada de Voltaje/fisiología , Arteria Retiniana/fisiología , 4-Aminopiridina/farmacología , Animales , Arteriolas , Electrofisiología , Endotelio Vascular , Masculino , Potenciales de la Membrana , Micotoxinas/farmacología , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio Calcio-Activados/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Arteria Retiniana/efectos de los fármacos , Arteria Retiniana/ultraestructura , Tetraetilamonio/farmacología
19.
Invest Ophthalmol Vis Sci ; 56(9): 5125-32, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26241400

RESUMEN

PURPOSE: Although L-type Ca2+ channels are known to play a key role in the myogenic reactivity of retinal arterial vessels, the involvement of other types of voltage-gated Ca2+ channels in this process remains unknown. In the present study we have investigated the contribution of T-type Ca2+ channels to myogenic signaling in arterioles of the rat retinal microcirculation. METHODS: Confocal immunolabeling of whole-mount preparations was used to investigate the localization of CaV3.1-3 channels in retinal arteriolar smooth muscle cells. T-type currents and the contribution of T-type channels to myogenic signaling were assessed by whole-cell patch-clamp recording and pressure myography of isolated retinal arteriole segments. RESULTS: Strong immunolabeling for CaV3.1 was observed on the plasma membrane of retinal arteriolar smooth muscle cells. In contrast, no expression of CaV3.2 or CaV3.3 could be detected in retinal arterioles, although these channels were present on glial cell end-feet surrounding the vessels and retinal ganglion cells, respectively. TTA-A2-sensitive T-type currents were recorded in retinal arteriolar myocytes with biophysical properties distinct from those of the L-type currents present in these cells. Inhibition of T-type channels using TTA-A2 or ML-218 dilated isolated, myogenically active, retinal arterioles. CONCLUSIONS: CaV3.1 T-type Ca2+ channels are functionally expressed on arteriolar smooth muscle cells of retinal arterioles and play an important role in myogenic signaling in these vessels. The work has important implications concerning our understanding of the mechanisms controlling blood flow autoregulation in the retina and its disruption during ocular disease.


Asunto(s)
Arteriolas/efectos de los fármacos , Bencenoacetamidas/farmacología , Músculo Liso Vascular/fisiología , Piridinas/farmacología , Vasos Retinianos/efectos de los fármacos , Vasoconstricción/fisiología , Animales , Inmunohistoquímica , Masculino , Potenciales de la Membrana/efectos de los fármacos , Técnicas de Placa-Clamp , Ratas , Vasos Retinianos/fisiología , Transducción de Señal/efectos de los fármacos , Vasoconstricción/efectos de los fármacos
20.
PLoS One ; 10(6): e0128359, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26047504

RESUMEN

Retinal endothelial cell dysfunction is believed to play a key role in the etiology and pathogenesis of diabetic retinopathy. Numerous studies have shown that TRPV4 channels are critically involved in maintaining normal endothelial cell function. In the current paper, we demonstrate that TRPV4 is functionally expressed in the endothelium of the retinal microcirculation and that both channel expression and activity is downregulated by hyperglycaemia. Quantitative PCR and immunostaining demonstrated molecular expression of TRPV4 in cultured bovine retinal microvascular endothelial cells (RMECs). Functional TRPV4 activity was assessed in cultured RMECs from endothelial Ca2+-responses recorded using fura-2 microfluorimetry and electrophysiological recordings of membrane currents. The TRPV4 agonist 4α-phorbol 12,13-didecanoate (4-αPDD) increased [Ca2+]i in RMECs and this response was largely abolished using siRNA targeted against TRPV4. These Ca2+-signals were completely inhibited by removal of extracellular Ca2+, confirming their dependence on influx of extracellular Ca2+. The 4-αPDD Ca2+-response recorded in the presence of cyclopiazonic acid (CPA), which depletes the intracellular stores preventing any signal amplification through store release, was used as a measure of Ca2+-influx across the cell membrane. This response was blocked by HC067047, a TRPV4 antagonist. Under voltage clamp conditions, the TRPV4 agonist GSK1016790A stimulated a membrane current, which was again inhibited by HC067047. Following incubation with 25 mM D-glucose TRPV4 expression was reduced in comparison with RMECs cultured under control conditions, as were 4αPDD-induced Ca2+-responses in the presence of CPA and ion currents evoked by GSK1016790A. Molecular expression of TRPV4 in the retinal vascular endothelium of 3 months' streptozotocin-induced diabetic rats was also reduced in comparison with that in age-matched controls. We conclude that hyperglycaemia and diabetes reduce the molecular and functional expression of TRPV4 channels in retinal microvascular endothelial cells. These changes may contribute to diabetes induced endothelial dysfunction and retinopathy.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Regulación hacia Abajo , Endotelio Vascular/metabolismo , Hiperglucemia/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Calcio/metabolismo , Bovinos , Células Cultivadas , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Endotelio Vascular/patología , Hiperglucemia/genética , Hiperglucemia/patología , Masculino , Microvasos/metabolismo , Microvasos/patología , Ratas Sprague-Dawley , Canales Catiónicos TRPV/análisis , Canales Catiónicos TRPV/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA